201
|
Li L, Chen J, Xiong G, St Clair DK, Xu W, Xu R. Increased ROS production in non-polarized mammary epithelial cells induces monocyte infiltration in 3D culture. J Cell Sci 2016; 130:190-202. [PMID: 27656113 DOI: 10.1242/jcs.186031] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 08/17/2016] [Indexed: 02/01/2023] Open
Abstract
Loss of epithelial cell polarity promotes cell invasion and cancer dissemination. Therefore, identification of factors that disrupt polarized acinar formation is crucial. Reactive oxygen species (ROS) drive cancer progression and promote inflammation. Here, we show that the non-polarized breast cancer cell line T4-2 generates significantly higher ROS levels than polarized S1 and T4R cells in three-dimensional (3D) culture, accompanied by induction of the nuclear factor κB (NF-κB) pathway and cytokine expression. Minimizing ROS in T4-2 cells with antioxidants reestablished basal polarity and inhibited cell proliferation. Introducing constitutively activated RAC1 disrupted cell polarity and increased ROS levels, indicating that RAC1 is a crucial regulator that links cell polarity and ROS generation. We also linked monocyte infiltration with disruption of polarized acinar structure using a 3D co-culture system. Gain- and loss-of-function experiments demonstrated that increased ROS in non-polarized cells is necessary and sufficient to enhance monocyte recruitment. ROS also induced cytokine expression and NF-κB activity. These results suggest that increased ROS production in mammary epithelial cell leads to disruption of cell polarity and promotes monocyte infiltration.
Collapse
Affiliation(s)
- Linzhang Li
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, Jilin Province 130021, China.,Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Jie Chen
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Gaofeng Xiong
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Daret K St Clair
- Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Wei Xu
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Ren Xu
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA .,Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
202
|
van den Bogaart E, Mens PF, Adams ER, Grobusch MP, Schallig HDFH. Phagocytosis of hemozoin by RAW 264.7 cells, but not THP-1 cells, promotes infection by Leishmania donovani with a nitric oxide-independent mechanism. Parasitol Int 2016; 66:196-206. [PMID: 27623326 DOI: 10.1016/j.parint.2016.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 08/19/2016] [Accepted: 09/08/2016] [Indexed: 01/28/2023]
Abstract
During its intra-erythrocytic development, the malaria parasite Plasmodium falciparum synthesizes insoluble hemozoin (HZ) crystals that are released into the circulation upon rupture of parasitized red blood cells, and rapidly phagocytized by host mononuclear cells. Here, HZ persists undigested, causing functional impairment and possibly leading to increased host susceptibility to secondary infections. In patients with malaria and visceral leishmaniasis (VL) co-infections, HZ-loaded macrophages are likely to co-harbor Leishmania donovani parasites, but whether this might influence the course of the Leishmania infection is unknown. In this study, L. donovani amastigote growth was monitored in mouse RAW 264.7 macrophages and PMA-differentiated THP-1 cells previously exposed to increasing amounts of HZ or its synthetic analogue β-hematin (BH). Latex beads were used as a phagocytic control. Data demonstrate that phagocytosis of HZ and BH by RAW 264.7 cells promoted infection therein by L. donovani parasites in a dose-dependent fashion. Similar results were not observed when using THP-1 cells, despite a clear persistence of undigested heme up to 48h after phagocytosis. Conditioning with lipopolysaccharide (LPS)/interferon (IFN)-γ prior to Leishmania infection triggered the release in RAW 264.7 cells of nitric oxide (NO), a highly leishmanicidal metabolite. However, neither HZ nor BH pre-ingestion were able to inhibit NO production following stimulation with LPS/IFN-γ, suggesting that the HZ- and BH-promoting effect on L. donovani infection occurred with an NO-independent mechanism. In conclusion, these preliminary findings highlight a possible detrimental effect of HZ on the course of VL, warranting further investigation into the clinical relevance of the current models.
Collapse
Affiliation(s)
- Erika van den Bogaart
- Parasitology Unit, Department of Biomedical Research, Royal Tropical Institute (KIT), Meibergdreef 39, 1105 AZ Amsterdam, The Netherlands.
| | - Pètra F Mens
- Parasitology Unit, Department of Medical Microbiology, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Emily R Adams
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Pl, Liverpool, Merseyside L3 5QA, United Kingdom
| | - Martin P Grobusch
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Henk D F H Schallig
- Parasitology Unit, Department of Medical Microbiology, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
203
|
Xiao D, Wang H, Han D. Single and combined genotoxicity effects of six pollutants on THP-1 cells. Food Chem Toxicol 2016; 95:96-102. [DOI: 10.1016/j.fct.2016.06.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/31/2016] [Accepted: 06/28/2016] [Indexed: 10/21/2022]
|
204
|
Betulin attenuates atherosclerosis in apoE -/- mice by up-regulating ABCA1 and ABCG1. Acta Pharmacol Sin 2016; 37:1337-1348. [PMID: 27374487 DOI: 10.1038/aps.2016.46] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/28/2016] [Indexed: 12/27/2022] Open
Abstract
AIM Betulin is a pentacyclic triterpenoid isolated from the bark of yellow and white birch trees with anti-cancer and anti-malaria activities. In this study we examined the effects of betulin on atherosclerosis in apoE-/- mice and the underlying mechanisms. METHODS Murine macrophage RAW264.7 cells and human monocyte-derived THP-1 cells were tested. Foam cell formation was detected with Oil Red O staining. Cholesterol efflux was assessed using [3H]-cholesterol efflux assay. The expression of ATP-binding cassette transporter A1 and G1 (ABCA1 and ABCG1) was examined using RT-PCR and Western-blotting. The ABCA1 promoter activity was evaluated using luciferase activity assay. Male apoE-/- mice fed on a high-fat-diet (HFD), and received betulin (20 and 40 mg·kg-1·d-1, ig) for 12 weeks. The macrophage content and ABCA1 expression in the aortic sinuses were evaluated with immunofluorescence staining. The hepatic, intestinal and fecal cholesterol were also analyzed in the mice. RESULTS In RAW264.7 cells, betulin (0.1-2.5 μg/mL) dose-dependently ameliorated oxLDL-induced cholesterol accumulation and enhanced cholesterol efflux. In both RAW264.7 and THP-1 cells, betulin increased the expression of ABCA1 and ABCG1 via suppressing the transcriptional repressors sterol-responsive element-binding proteins (SREBPs) that bound to E-box motifs in ABCA1 promoter, whereas E-box binding site mutation markedly attenuated betulin-induced ABCA1 promoter activity. In HFD-fed apoE-/- mice, betulin administration significantly reduced lesions in en face aortas and aortic sinuses. Furthermore, betulin administration significantly increased ABCA1 expression and suppressed macrophage positive areas in the aortic sinuses. Moreover, betulin administration improved plasma lipid profiles and enhanced fecal cholesterol excretion in the mice. CONCLUSION Betulin attenuates atherosclerosis in apoE-/- mice by promoting cholesterol efflux in macrophages.
Collapse
|
205
|
Nouman M, Jubeli E, Saunier J, Yagoubi N. Exudation of additives to the surface of medical devices: impact on biocompatibility in the case of polyurethane used in implantable catheters. J Biomed Mater Res A 2016; 104:2954-2967. [DOI: 10.1002/jbm.a.35837] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/21/2016] [Indexed: 01/08/2023]
Affiliation(s)
- Micheal Nouman
- Université Paris-Sud, EA401 Matériaux et Santé, IFR 141, Faculté De Pharmacie; Châtenay Malabry France
| | - Emile Jubeli
- Université Paris-Sud, EA401 Matériaux et Santé, IFR 141, Faculté De Pharmacie; Châtenay Malabry France
| | - Johanna Saunier
- Université Paris-Sud, EA401 Matériaux et Santé, IFR 141, Faculté De Pharmacie; Châtenay Malabry France
| | - Najet Yagoubi
- Université Paris-Sud, EA401 Matériaux et Santé, IFR 141, Faculté De Pharmacie; Châtenay Malabry France
| |
Collapse
|
206
|
Delannoy CP, Rombouts Y, Groux-Degroote S, Holst S, Coddeville B, Harduin-Lepers A, Wuhrer M, Elass-Rochard E, Guérardel Y. Glycosylation Changes Triggered by the Differentiation of Monocytic THP-1 Cell Line into Macrophages. J Proteome Res 2016; 16:156-169. [DOI: 10.1021/acs.jproteome.6b00161] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Clément P. Delannoy
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité
de Glycobiologie Structurale et Fonctionnelle, F 59000 Lille, France
| | - Yoann Rombouts
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité
de Glycobiologie Structurale et Fonctionnelle, F 59000 Lille, France
| | - Sophie Groux-Degroote
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité
de Glycobiologie Structurale et Fonctionnelle, F 59000 Lille, France
| | - Stephanie Holst
- Center
for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Bernadette Coddeville
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité
de Glycobiologie Structurale et Fonctionnelle, F 59000 Lille, France
| | - Anne Harduin-Lepers
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité
de Glycobiologie Structurale et Fonctionnelle, F 59000 Lille, France
| | - Manfred Wuhrer
- Center
for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Elisabeth Elass-Rochard
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité
de Glycobiologie Structurale et Fonctionnelle, F 59000 Lille, France
| | - Yann Guérardel
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité
de Glycobiologie Structurale et Fonctionnelle, F 59000 Lille, France
| |
Collapse
|
207
|
Futakuchi A, Inoue T, Fujimoto T, Inoue-Mochita M, Kawai M, Tanihara H. The effects of ripasudil (K-115), a Rho kinase inhibitor, on activation of human conjunctival fibroblasts. Exp Eye Res 2016; 149:107-115. [PMID: 27394186 DOI: 10.1016/j.exer.2016.07.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 06/17/2016] [Accepted: 07/05/2016] [Indexed: 02/06/2023]
Abstract
The most common cause of glaucoma surgery failure is scar formation induced by activation of wound-healing responses and resultant fibrosis at the surgical site. We investigated the effects of ripasudil, a Rho kinase inhibitor, on activation of human conjunctival fibroblasts (HConF). HConF were pretreated with different concentrations of ripasudil for 1 h before addition of transforming growth factor (TGF)-β2, followed by incubation for 48 h. TGF-β2-treated fibroblasts exhibited a significant increase in expression of α-smooth muscle actin (α-SMA), a marker of fibroblast-to-myofibroblast differentiation, and this increase was significantly suppressed, in a dose-dependent manner, by pretreatment with ripasudil. Ripasudil pretreatment also significantly attenuated TGF-β2-induced fibronectin production and collagen gel contraction. TGF-β2 increased both the number of viable cells and the number of cells in the G2/M phase of the cell cycle; these effects were attenuated by pretreatment with ripasudil. In addition, we explored the effects of ripasudil on stimulation of HConF by activated macrophages. Human monocytic cell line THP-1 cells were differentiated into M1 or M2 macrophage-like cells, and HConF were treated with conditioned media derived from these macrophages in the presence or absence of ripasudil. Conditioned medium from M2 macrophage-like cells induced a significant increase in α-SMA expression, viable cell numbers, and gel contraction, all of which were significantly suppressed by ripasudil. Thus, overall, ripasudil attenuated activation of human conjunctival fibroblasts. Ripasudil may be of therapeutic utility, preventing excessive scarring after glaucoma filtration surgery.
Collapse
Affiliation(s)
- Akiko Futakuchi
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Toshihiro Inoue
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| | - Tomokazu Fujimoto
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Miyuki Inoue-Mochita
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Motofumi Kawai
- Department of Ophthalmology, Asahikawa Medical University, Asahikawa, Japan
| | - Hidenobu Tanihara
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
208
|
Tucureanu MM, Butoi E, Gan AM, Stan D, Constantinescu CA, Calin M, Simionescu M, Manduteanu I. Amendment of the cytokine profile in macrophages subsequent to their interaction with smooth muscle cells: Differential modulation by fractalkine and resistin. Cytokine 2016; 83:250-261. [DOI: 10.1016/j.cyto.2016.04.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 04/28/2016] [Accepted: 04/28/2016] [Indexed: 11/26/2022]
|
209
|
Pelin M, Florio C, Ponti C, Lucafò M, Gibellini D, Tubaro A, Sosa S. Pro-inflammatory effects of palytoxin: an in vitro study on human keratinocytes and inflammatory cells. Toxicol Res (Camb) 2016; 5:1172-1181. [PMID: 30090423 PMCID: PMC6060726 DOI: 10.1039/c6tx00084c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 05/13/2016] [Indexed: 12/14/2022] Open
Abstract
Palytoxin (PLTX) is one of the most harmful marine toxins known so far. Although the ingestion of contaminated seafood is the most dangerous exposure route for humans, cutaneous and inhalational exposures are far more frequent, and can cause strong inflammatory reactions. However, little is known about the inflammatory events that follow the cutaneous exposure to the toxin. In this study, we investigated (1) the effects of both short (2 h) and long (24 h) term exposures of HaCaT keratinocytes to a sub-cytotoxic PLTX concentration on pro-inflammatory mediator gene expression and release and (2) the effect of PLTX-conditioned HaCaT cell media on undifferentiated (monocytes) and differentiated (macrophages; immature dendritic cells, iDCs; mature dendritic cells, mDCs) THP-1 cells. At 10-11 M, PLTX induced interleukin (IL)-6 and IL-8 release from HaCaT keratinocytes after 24 h of continuous exposure to the toxin, as well as after 23 h in toxin-free medium preceded by 1 h exposure to PLTX. Under the same experimental conditions, release of the inflammatory mediators prostaglandin-E2 and histamine was also found after both short and long exposures to the toxin. The conditioned media collected from HaCaT cells treated with PLTX increased the migration of the differentiated and undifferentiated THP-1 cells (potency rank order: monocytes ≥ iDCs > mDCs > macrophages) but did not induce cell differentiation. These results indicate that keratinocytes can be actively involved in the recruitment of inflammatory cells in response to cutaneous contact with PLTX. The lack of a significant effect on monocyte differentiation towards mature immune cells suggests that PLTX is endowed with irritant rather than sensitizing properties.
Collapse
Affiliation(s)
- Marco Pelin
- Department of Life Sciences , University of Trieste , 34127 Trieste , Italy . ; Tel: +39-040-5588835
| | - Chiara Florio
- Department of Life Sciences , University of Trieste , 34127 Trieste , Italy . ; Tel: +39-040-5588835
| | - Cristina Ponti
- Department of Life Sciences , University of Trieste , 34127 Trieste , Italy . ; Tel: +39-040-5588835
| | - Marianna Lucafò
- Department of Medical , Surgical and Health Sciences , University of Trieste , 34127 Trieste , Italy
| | - Davide Gibellini
- Department of Pathology and Diagnostic , University of Verona , 35124 Verona , Italy
| | - Aurelia Tubaro
- Department of Life Sciences , University of Trieste , 34127 Trieste , Italy . ; Tel: +39-040-5588835
| | - Silvio Sosa
- Department of Life Sciences , University of Trieste , 34127 Trieste , Italy . ; Tel: +39-040-5588835
| |
Collapse
|
210
|
O'Connor G, Gleeson LE, Fagan-Murphy A, Cryan SA, O'Sullivan MP, Keane J. Sharpening nature's tools for efficient tuberculosis control: A review of the potential role and development of host-directed therapies and strategies for targeted respiratory delivery. Adv Drug Deliv Rev 2016; 102:33-54. [PMID: 27151307 DOI: 10.1016/j.addr.2016.04.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 04/04/2016] [Accepted: 04/20/2016] [Indexed: 12/18/2022]
Abstract
Centuries since it was first described, tuberculosis (TB) remains a significant global public health issue. Despite ongoing holistic measures implemented by health authorities and a number of new oral treatments reaching the market, there is still a need for an advanced, efficient TB treatment. An adjunctive, host-directed therapy designed to enhance endogenous pathways and hence compliment current regimens could be the answer. The integration of drug repurposing, including synthetic and naturally occurring compounds, with a targeted drug delivery platform is an attractive development option. In order for a new anti-tubercular treatment to be produced in a timely manner, a multidisciplinary approach should be taken from the outset including stakeholders from academia, the pharmaceutical industry, and regulatory bodies keeping the patient as the key focus. Pre-clinical considerations for the development of a targeted host-directed therapy are discussed here.
Collapse
Affiliation(s)
- Gemma O'Connor
- School of Pharmacy, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin and St. James's Hospital, D08 W9RT, Dublin, Ireland.
| | - Laura E Gleeson
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin and St. James's Hospital, D08 W9RT, Dublin, Ireland.
| | - Aidan Fagan-Murphy
- School of Pharmacy, Royal College of Surgeons in Ireland, Dublin 2, Ireland; SFI Centre for Research in Medical Devices (CURAM), Dublin 2, Ireland.
| | - Sally-Ann Cryan
- School of Pharmacy, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Trinity Centre for Bioengineering, Trinity College Dublin, Dublin 2, Ireland; SFI Centre for Research in Medical Devices (CURAM), Dublin 2, Ireland.
| | - Mary P O'Sullivan
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin and St. James's Hospital, D08 W9RT, Dublin, Ireland.
| | - Joseph Keane
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin and St. James's Hospital, D08 W9RT, Dublin, Ireland.
| |
Collapse
|
211
|
Novel NSAID-Derived Drugs for the Potential Treatment of Alzheimer's Disease. Int J Mol Sci 2016; 17:ijms17071035. [PMID: 27376271 PMCID: PMC4964411 DOI: 10.3390/ijms17071035] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 06/13/2016] [Accepted: 06/20/2016] [Indexed: 12/13/2022] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) have been suggested for the potential treatment of neurodegenerative diseases, such as Alzheimer's disease (AD). Prolonged use of NSAIDs, however, produces gastrointestinal (GI) toxicity. To overcome this serious limitation, the aim of this study was to develop novel NSAID-derived drug conjugates (Anti-inflammatory-Lipoyl derivatives, AL4-9) that preserve the beneficial effects of NSAIDS without causing GI problems. As such, we conjugated selected well-known NSAIDs, such as (S)-naproxen and (R)-flurbiprofen, with (R)-α-lipoic acid (LA) through alkylene diamine linkers. The selection of the antioxidant LA was based on the proposed role of oxidative stress in the development and/or progression of AD. Our exploratory studies revealed that AL7 containing the diaminoethylene linker between (R)-flurbiprofen and LA had the most favorable chemical and in vitro enzymatic stability profiles among the synthesized compounds. Upon pretreatment, this compound exhibited excellent antioxidant activity in phorbol 12-miristate 13-acetate (PMA)-stimulated U937 cells (lymphoblast lung from human) and Aβ(25-35)-treated THP-1 cells (leukemic monocytes). Furthermore, AL7 also modulated the expression of COX-2, IL-1β and TNF-α in these cell lines, suggesting anti-inflammatory activity. Taken together, AL7 has emerged as a potential lead worthy of further characterization and testing in suitable in vivo models of AD.
Collapse
|
212
|
Monocyte Differentiation towards Protumor Activity Does Not Correlate with M1 or M2 Phenotypes. J Immunol Res 2016; 2016:6031486. [PMID: 27376091 PMCID: PMC4916292 DOI: 10.1155/2016/6031486] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/04/2016] [Indexed: 11/17/2022] Open
Abstract
Macrophages facilitate breast cancer progression. Macrophages were initially classified as M1 or M2 based on their distinct metabolic programs and then expanded to include antitumoral (M1) and protumoral (M2) activities. However, it is still uncertain what markers define the pro- and antitumoral phenotypes and what conditions lead to their formation. In this study, monocytic cell lines and primary monocytes were subjected to commonly reported protocols of M1/M2 polarization and conditions known to engage monocytes into protumoral functions. The results showed that only IDO enzyme and CD86 M1 markers were upregulated correlating with M1 polarization. TNF-α, CCR7, IL-10, arginase I, CD36, and CD163 were expressed indistinguishably from M1 or M2 polarization. Similarly, protumoral engaging resulted in upregulation of both M1 and M2 markers, with conditioned media from the most aggressive breast cancer cell line promoting the greatest changes. In spite of the mixed phenotype, M1-polarized macrophages exhibited the highest expression/secretion of inflammatory mediators, many of which have previously been associated with breast cancer aggressiveness. These data argue that although the existence of protumoral macrophages is unquestionable, their associated phenotypes and the precise conditions driving their formation are still unclear, and those conditions may need both M1 and M2 stimuli.
Collapse
|
213
|
Tarasova NK, Ytterberg AJ, Lundberg K, Zhang XM, Harris RA, Zubarev RA. Establishing a Proteomics-Based Monocyte Assay To Assess Differential Innate Immune Activation Responses. J Proteome Res 2016; 15:2337-45. [PMID: 27223872 DOI: 10.1021/acs.jproteome.6b00422] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Innate immune cells are complex systems that can be simultaneously activated in a variety of ways. Common methods currently used to estimate the response of innate immune cells to stimuli are usually biased toward a single mode of activation. The aim of this study was to assess the possibility of designing an assay based on unbiased proteome analysis that would be capable of predicting the complex response of the innate immune system to various challenges. Monocytes were used as representative cells of the innate immune system. The underlying hypothesis was that their proteome response to different activating molecules would reflect the immunogenicity of these molecules. To identify the main modes of response, we treated the human monocytic THP-1 cell line with nine different stimuli. Differentiation and activation were determined to be the two major modes of monocyte response, with PMA causing the strongest differentiation and Pam3CSK4 causing the strongest proinflammatory activation. The established assay was applied to characterize the monocyte response to epidermal growth factor peptide containing isoaspartate, which induced differentiation but not proinflammatory activation. Because of its versatility, robustness, and specificity, this new assay is likely to find a niche among the more established immunological methods.
Collapse
Affiliation(s)
| | | | | | - Xing-Mei Zhang
- Department of Clinical Neuroscience, Centre for Molecular Medicine, Karolinska Hospital, Karolinska Institutet , SE 17176 Stockholm, Sweden
| | - Robert A Harris
- Department of Clinical Neuroscience, Centre for Molecular Medicine, Karolinska Hospital, Karolinska Institutet , SE 17176 Stockholm, Sweden
| | | |
Collapse
|
214
|
Sulforaphane regulates phenotypic and functional switching of both induced and spontaneously differentiating human monocytes. Int Immunopharmacol 2016; 35:85-98. [DOI: 10.1016/j.intimp.2016.03.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 03/05/2016] [Accepted: 03/08/2016] [Indexed: 12/26/2022]
|
215
|
Foit L, Thaxton CS. Synthetic high-density lipoprotein-like nanoparticles potently inhibit cell signaling and production of inflammatory mediators induced by lipopolysaccharide binding Toll-like receptor 4. Biomaterials 2016; 100:67-75. [PMID: 27244690 DOI: 10.1016/j.biomaterials.2016.05.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 05/15/2016] [Accepted: 05/19/2016] [Indexed: 12/25/2022]
Abstract
Toll-like receptor 4 (TLR4) plays a critical role in the innate immune system. Stimulation of TLR4 occurs upon binding lipopolysaccharide (LPS), a component of Gram-negative bacterial cell walls. Due to the potency of the induced inflammatory response, there is a growing interest in agents that can most proximally modulate this LPS/TLR4 interaction to prevent downstream cell signaling events and the production of inflammatory mediators. Building on the natural ability of human high-density lipoprotein (HDL) to bind LPS, we synthesized a suite of HDL-like nanoparticles (HDL-like NP). We identified one HDL-like NP that was particularly effective at decreasing TLR4 signaling caused by addition of purified LPS or Gram-negative bacteria to model human cell lines or primary human peripheral blood cells. The HDL-like NP functioned to inhibit TLR4-dependent inflammatory response to LPS derived from multiple bacterial species. Mechanistically, data show that the NP mainly functions by scavenging and neutralizing the LPS toxin. Taken together, HDL-like NPs constitute a powerful endotoxin scavenger with the potential to significantly reduce LPS-mediated inflammation.
Collapse
Affiliation(s)
- Linda Foit
- Feinberg School of Medicine, Department of Urology, Northwestern University, Tarry 16-703, 303 E. Chicago Ave, Chicago, IL 60611, USA; Simpson Querrey Institute for BioNanotechnology, Northwestern University, 303 E. Superior St, Chicago, IL 60611, USA
| | - C Shad Thaxton
- Feinberg School of Medicine, Department of Urology, Northwestern University, Tarry 16-703, 303 E. Chicago Ave, Chicago, IL 60611, USA; Simpson Querrey Institute for BioNanotechnology, Northwestern University, 303 E. Superior St, Chicago, IL 60611, USA; International Institute for Nanotechnology (IIN), 2145 Sheridan Road, Evanston, IL 60208, USA; Robert H Lurie Comprehensive Cancer Center (RHLCCC), Northwestern University, Feinberg School of Medicine, 303 E Superior, Chicago, IL 60611, USA.
| |
Collapse
|
216
|
Bonifati S, Daly MB, St Gelais C, Kim SH, Hollenbaugh JA, Shepard C, Kennedy EM, Kim DH, Schinazi RF, Kim B, Wu L. SAMHD1 controls cell cycle status, apoptosis and HIV-1 infection in monocytic THP-1 cells. Virology 2016; 495:92-100. [PMID: 27183329 DOI: 10.1016/j.virol.2016.05.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 05/03/2016] [Indexed: 01/09/2023]
Abstract
SAMHD1 limits HIV-1 infection in non-dividing myeloid cells by decreasing intracellular dNTP pools. HIV-1 restriction by SAMHD1 in these cells likely prevents activation of antiviral immune responses and modulates viral pathogenesis, thus highlighting a critical role of SAMHD1 in HIV-1 physiopathology. Here, we explored the function of SAMHD1 in regulating cell proliferation, cell cycle progression and apoptosis in monocytic THP-1 cells. Using the CRISPR/Cas9 technology, we generated THP-1 cells with stable SAMHD1 knockout. We found that silencing of SAMHD1 in cycling cells stimulates cell proliferation, redistributes cell cycle population in the G1/G0 phase and reduces apoptosis. These alterations correlated with increased dNTP levels and more efficient HIV-1 infection in dividing SAMHD1 knockout cells relative to control. Our results suggest that SAMHD1, through its dNTPase activity, affects cell proliferation, cell cycle distribution and apoptosis, and emphasize a key role of SAMHD1 in the interplay between cell cycle regulation and HIV-1 infection.
Collapse
Affiliation(s)
- Serena Bonifati
- Center for Retrovirus Research, Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Michele B Daly
- Center for Drug Discovery, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Corine St Gelais
- Center for Retrovirus Research, Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Sun Hee Kim
- Center for Retrovirus Research, Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Joseph A Hollenbaugh
- Center for Drug Discovery, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Caitlin Shepard
- Center for Drug Discovery, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Edward M Kennedy
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Dong-Hyun Kim
- Department of Pharmacy, School of Pharmacy, Kyung-Hee University, Seoul, South Korea
| | - Raymond F Schinazi
- Center for Drug Discovery, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Baek Kim
- Center for Drug Discovery, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA; Department of Pharmacy, School of Pharmacy, Kyung-Hee University, Seoul, South Korea.
| | - Li Wu
- Center for Retrovirus Research, Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
217
|
Fazalul Rahiman SS, Morgan M, Gray P, Shaw PN, Cabot PJ. Dynorphin 1-17 and Its N-Terminal Biotransformation Fragments Modulate Lipopolysaccharide-Stimulated Nuclear Factor-kappa B Nuclear Translocation, Interleukin-1beta and Tumor Necrosis Factor-alpha in Differentiated THP-1 Cells. PLoS One 2016; 11:e0153005. [PMID: 27055013 PMCID: PMC4824444 DOI: 10.1371/journal.pone.0153005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/21/2016] [Indexed: 12/31/2022] Open
Abstract
Dynorphin 1–17, (DYN 1–17) opioid peptide produces antinociception following binding to the kappa-opioid peptide (KOP) receptor. Upon synthesis and release in inflamed tissues by immune cells, DYN 1–17 undergoes rapid biotransformation and yields a unique set of opioid and non-opioid fragments. Some of these major fragments possess a role in immunomodulation, suggesting that opioid-targeted therapeutics may be effective in diminishing the severity of inflammatory disorders. This study aimed to examine the immunomodulatory effects of DYN 1–17 and major N-terminal fragments found in the inflammatory environment on nuclear factor-kappaB/p65 (NF-κB/p65) nuclear translocation and the release of interleukin-1beta (IL-1β) and tumor necrosis factor-alpha (TNF-α) from lipopolysaccharide (LPS)-stimulated, differentiated THP-1 cells. The results demonstrate that NF-κB/p65 nuclear translocation was significantly attenuated following treatment with DYN 1–17 and a specific range of fragments, with the greatest reduction observed with DYN 1–7 at a low concentration (10 nM). Antagonism with a selective KOP receptor antagonist, ML-190, significantly reversed the inhibitory effects of DYN 1–17, DYN 1–6, DYN 1–7 and DYN 1–9, but not other DYN 1–17 N-terminal fragments (DYN 1–10 and 1–11) on NF-κB/p65 nuclear translocation. DYN 1–17 and selected fragments demonstrated differential modulation on the release of IL-1β and TNF-α with significant inhibition observed with DYN 1–7 at low concentrations (1 nM and 10 pM). These effects were blocked by ML-190, suggesting a KOP receptor-mediated pathway. The results demonstrate that DYN 1–17 and certain N-terminal fragments, produced in an inflamed environment, play an anti-inflammatory role by inhibiting NF-κB/p65 translocation and the subsequent cytokine release through KOP receptor-dependent and independent pathways.
Collapse
Affiliation(s)
- Siti Sarah Fazalul Rahiman
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Michael Morgan
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Paul Gray
- School of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Department of Anaesthesia, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Paul Nicholas Shaw
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
| | - Peter John Cabot
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
- * E-mail:
| |
Collapse
|
218
|
Sharma NK, Sajuthi SP, Chou JW, Calles-Escandon J, Demons J, Rogers S, Ma L, Palmer ND, McWilliams DR, Beal J, Comeau ME, Cherry K, Hawkins GA, Menon L, Kouba E, Davis D, Burris M, Byerly SJ, Easter L, Bowden DW, Freedman BI, Langefeld CD, Das SK. Tissue-Specific and Genetic Regulation of Insulin Sensitivity-Associated Transcripts in African Americans. J Clin Endocrinol Metab 2016; 101:1455-68. [PMID: 26789776 PMCID: PMC4880154 DOI: 10.1210/jc.2015-3336] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Integrative multiomics analyses of adipose and muscle tissue transcripts, S, and genotypes revealed novel genetic regulatory mechanisms of insulin resistance in African Americans.
Collapse
Affiliation(s)
- Neeraj K Sharma
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Satria P Sajuthi
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Jeff W Chou
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Jorge Calles-Escandon
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Jamehl Demons
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Samantha Rogers
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Lijun Ma
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Nicholette D Palmer
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - David R McWilliams
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - John Beal
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Mary E Comeau
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Kristina Cherry
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Gregory A Hawkins
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Lata Menon
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Ethel Kouba
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Donna Davis
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Marcie Burris
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Sara J Byerly
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Linda Easter
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Donald W Bowden
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Barry I Freedman
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Carl D Langefeld
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Swapan K Das
- Department of Internal Medicine (N.K.S., J.C.-E., J.D., S.R., L.Ma., K.C., L.Me., E.K., D.D., B.I.F., S.K.D.), Center for Public Health Genomics (N.K.S., S.P.S., J.W.C., L.Ma., N.D.P., D.R.M., M.C., G.A.H., B.I.F., C.D.L., S.K.D.), Department of Biostatistical Sciences, Division of Public Health Sciences (S.P.S., J.W.C., D.R.M., J.B., M.C., C.D.L.), Department of Biochemistry (N.D.P., D.W.B.), Center for Diabetes Research and Center for Genomics and Personalized Medicine Research (N.D.P., G.A.H., D.W.B., B.I.F.), and Clinical Research Unit, Biomedical Research Services and Administration (M.B., S.J.B., L.E.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| |
Collapse
|
219
|
Richter E, Ventz K, Harms M, Mostertz J, Hochgräfe F. Induction of Macrophage Function in Human THP-1 Cells Is Associated with Rewiring of MAPK Signaling and Activation of MAP3K7 (TAK1) Protein Kinase. Front Cell Dev Biol 2016; 4:21. [PMID: 27066479 PMCID: PMC4811913 DOI: 10.3389/fcell.2016.00021] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/09/2016] [Indexed: 12/12/2022] Open
Abstract
Macrophages represent the primary human host response to pathogen infection and link the immediate defense to the adaptive immune system. Mature tissue macrophages convert from circulating monocyte precursor cells by terminal differentiation in a process that is not fully understood. Here, we analyzed the protein kinases of the human monocytic cell line THP-1 before and after induction of macrophage differentiation by using kinomics and phosphoproteomics. When comparing the macrophage-like state with the monocytic precursor, 50% of the kinome was altered in expression and even 71% of covered kinase phosphorylation sites were affected. Kinome rearrangements are for example characterized by a shift of overrepresented cyclin-dependent kinases associated with cell cycle control in monocytes to calmodulin-dependent kinases and kinases involved in proinflammatory signaling. Eventually, we show that monocyte-to-macrophage differentiation is associated with major rewiring of mitogen-activated protein kinase signaling networks and demonstrate that protein kinase MAP3K7 (TAK1) acts as the key signaling hub in bacterial killing, chemokine production and differentiation. Our study proves the fundamental role of protein kinases and cellular signaling as major drivers of macrophage differentiation and function. The finding that MAP3K7 is central to macrophage function suggests MAP3K7 and its networking partners as promising targets in host-directed therapy for macrophage-associated disease.
Collapse
Affiliation(s)
- Erik Richter
- Junior Research Group Pathoproteomics, Competence Center Functional Genomics, University of Greifswald Greifswald, Germany
| | - Katharina Ventz
- Junior Research Group Pathoproteomics, Competence Center Functional Genomics, University of Greifswald Greifswald, Germany
| | - Manuela Harms
- Junior Research Group Pathoproteomics, Competence Center Functional Genomics, University of Greifswald Greifswald, Germany
| | - Jörg Mostertz
- Junior Research Group Pathoproteomics, Competence Center Functional Genomics, University of Greifswald Greifswald, Germany
| | - Falko Hochgräfe
- Junior Research Group Pathoproteomics, Competence Center Functional Genomics, University of Greifswald Greifswald, Germany
| |
Collapse
|
220
|
Millrine D, Miyata H, Tei M, Dubey P, Nyati K, Nakahama T, Gemechu Y, Ripley B, Kishimoto T. Immunomodulatory drugs inhibit TLR4-induced type-1 interferon production independently of Cereblon via suppression of the TRIF/IRF3 pathway. Int Immunol 2016; 28:307-15. [PMID: 26865412 DOI: 10.1093/intimm/dxw005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/07/2016] [Indexed: 12/29/2022] Open
Abstract
Thalidomide and its derivatives, collectively referred to as immunomodulatory drugs (IMiDs), are effective inhibitors of inflammation and are known to inhibit TLR-induced TNFα production. The identification of Cereblon as the receptor for these compounds has led to a rapid advancement in our understanding of IMiD properties; however, there remain no studies addressing the role of Cereblon in mediating the suppressive effect of IMiDs on TLR responses. Here, we developed Cereblon-deficient mice using the CRISPR-Cas9 system. TLR-induced cytokine responses were unaffected by Cereblon deficiency in vivo Moreover, IMiD treatment inhibited cytokine production even in the absence of Cereblon. The IMiD-induced suppression of cytokine production therefore occurs independently of Cereblon in mice. Further investigation revealed that IMiDs are potent inhibitors of TLR-induced type-1 interferon production via suppression of the TRIF/IRF3 pathway. These data suggest that IMiDs may prove effective in the treatment of disorders characterized by the ectopic production of type-1 interferon. Significantly, these properties are mediated separately from thalidomide's teratogenic receptor, Cereblon. Thus, certain therapeutic properties of Thalidomide can be separated from its harmful side effects.
Collapse
Affiliation(s)
- David Millrine
- Laboratory of Immune Regulation, IFReC Research Building, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Haruhiko Miyata
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Mami Tei
- Laboratory of Immune Regulation, IFReC Research Building, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Praveen Dubey
- Laboratory of Immune Regulation, IFReC Research Building, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kishan Nyati
- Laboratory of Immune Regulation, IFReC Research Building, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Taisuke Nakahama
- Laboratory of Immune Regulation, IFReC Research Building, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yohannes Gemechu
- Laboratory of Immune Regulation, IFReC Research Building, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Barry Ripley
- Laboratory of Immune Regulation, IFReC Research Building, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tadamitsu Kishimoto
- Laboratory of Immune Regulation, IFReC Research Building, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
221
|
He W, Frueh J, Wu Z, He Q. How Leucocyte Cell Membrane Modified Janus Microcapsules are Phagocytosed by Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2016; 8:4407-4415. [PMID: 26824329 DOI: 10.1021/acsami.5b10885] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Modern drug delivery systems rely on either antibody-based single-surface recognition or on surface-hydrophobicity-based approaches. For a tumor showing various surface mutations, both approaches fail. This publication hereby presents Janus capsules based on polyelectrolyte multilayer microcapsules exhibiting human leucocyte (THP-1 cell line) cell membranes for discriminating HUVEC cells from three different cancer cell lines. Despite destroying the cellular integrity of leucocyte cells, the modified Janus capsules are able to adhere to cancer cells. Leucocyte cell-membrane-coated Janus capsules are phagocytosed with the cellular membrane part pointing to the cells.
Collapse
Affiliation(s)
- Wenping He
- Key Laboratory of Microsystems and Microstructures Manufacturing, Ministry of Education, Micro/Nano Technology Research Centre, Harbin Institute of Technology , Yikuang Street 2 B1, Harbin 150080, China
| | - Johannes Frueh
- Key Laboratory of Microsystems and Microstructures Manufacturing, Ministry of Education, Micro/Nano Technology Research Centre, Harbin Institute of Technology , Yikuang Street 2 B1, Harbin 150080, China
| | - Zhenwei Wu
- Key Laboratory of Microsystems and Microstructures Manufacturing, Ministry of Education, Micro/Nano Technology Research Centre, Harbin Institute of Technology , Yikuang Street 2 B1, Harbin 150080, China
| | - Qiang He
- Key Laboratory of Microsystems and Microstructures Manufacturing, Ministry of Education, Micro/Nano Technology Research Centre, Harbin Institute of Technology , Yikuang Street 2 B1, Harbin 150080, China
| |
Collapse
|
222
|
The choice of phorbol 12-myristate 13-acetate differentiation protocol influences the response of THP-1 macrophages to a pro-inflammatory stimulus. J Immunol Methods 2016; 430:64-70. [PMID: 26826276 DOI: 10.1016/j.jim.2016.01.012] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 01/04/2016] [Accepted: 01/26/2016] [Indexed: 11/22/2022]
Abstract
The human monocytic cell line, THP-1, is the most widely used model for primary human monocytes/macrophages. This is because, following differentiation using phorbol 12-myristate 13-acetate (PMA), THP-1 cells acquire a macrophage-like phenotype, which mimics, in many respects, primary human macrophages. Despite the widespread use of THP-1 cells in studies elucidating macrophage responses to inflammatory stimuli, as well as the development and screening of potential therapeutics, there is currently no standardised protocol for the reliable differentiation of THP-1 monocytes to a macrophage phenotype using PMA. Consequently, reports using THP-1 cells have demonstrated significant phenotypic and functional differences between resultant THP-1 macrophage populations, which are largely attributable to the varying PMA differentiation methods used. Thus, to guarantee consistency and reproducibility between studies, and to ensure the relevance of THP-1 cells as an appropriate model for primary human macrophages, it is crucial to develop a standardised protocol for the differentiation of THP-1 macrophages. Accordingly, we compared the function and phenotype of THP-1 macrophages generated using the range of published PMA differentiation protocols, specifically in response to the pro-inflammatory stimulus, lipopolysaccharide (LPS). Our results demonstrated that the function of the resultant THP-1 macrophage populations, as determined by tumour necrosis factor (TNF) secretion in response to LPS stimulation, varied significantly, and was dependent upon the concentration of PMA used to stimulate the differentiation of monocytes, and the period of rest following PMA exposure. These data indicate that exposure of monocytic THP-1 cells to 25 nM PMA over 48 h, followed by a recovery period of 24h in culture in the absence of PMA, was the optimal protocol for the differentiation of THP-1 cells.
Collapse
|
223
|
A novel miR-200b-3p/p38IP pair regulates monocyte/macrophage differentiation. Cell Discov 2016; 2:15043. [PMID: 27462440 PMCID: PMC4860955 DOI: 10.1038/celldisc.2015.43] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 11/05/2015] [Indexed: 01/13/2023] Open
Abstract
Monocyte/macrophage differentiation represents a major branch of hematopoiesis and is a central event in the immune response, but the molecular mechanisms underlying are not fully delineated. Here we show that p38 mitogen-activated protein kinase (MAPK) interacting protein (p38IP) is downregulated during monocyte/macrophage differentiation in vitro. Overexpression of p38IP halted monocyte/macrophage differentiation, whereas forward knockdown of p38IP by RNA interference induced G1/S arrest and promoted monocyte differentiation into macrophages and the maturation of macrophages as well. Moreover, we found that miR-200b-3p was upregulated during monocyte/macrophage differentiation and mediated the downregulation of p38IP by binding to the 3′ untranslated terminal region of p38IP mRNA. Overexpression of a miR-200b-3p mimic resembled the effect of p38IP knockdown, whereas a miR-200b-3p inhibitor blocked monocyte/macrophage differentiation by enhancing p38IP expression. Further western blotting analysis revealed that p38IP downregulation enhanced the activity of p38 MAPK and the subsequent accumulation of cyclin-dependent kinase inhibitor p21, thus promoting G1/S arrest and monocyte/macrophage differentiation. Moreover, the decline of GCN5 acetyltransferase caused by p38IP downregulation was required but was not sufficient for monocyte/macrophage differentiation. This study demonstrated a new role for p38IP and a novel miR-200b-3p/p38IP pair in the regulation of monocyte/macrophage differentiation.
Collapse
|
224
|
Zeng J, Chen QW, Yu ZY, Zhang JR, Chen DL, Song C, Luo J, Zhang C, Wang SL, Chen JP. Regulation of intrinsic apoptosis in cycloheximide-treated macrophages by the Sichuan human strain of Chinese Leishmania isolates. Acta Trop 2016; 153:101-10. [PMID: 26482137 DOI: 10.1016/j.actatropica.2015.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 08/27/2015] [Accepted: 10/12/2015] [Indexed: 10/22/2022]
Abstract
Leishmania spp. are able to survive and proliferate inside mammals' mononuclear phagocytes, causing Leishmaniasis. Previous studies have noted that the regulation of apoptosis in host cells by these parasites may contribute to their ability to evade the immune system. However, current results remain unclear about whether the parasites can promote or delay the apoptotic process in host cells, because the regulatory effect of Leishmania was assumed to be strain-, species- and even infection time-dependent. The aim of this study was to investigate whether the Sichuan isolates of Chinese Leishmania (SC10H2) can alter the process of intrinsic apoptosis induced by cycloheximide in different types of macrophage cell lines and to determine in which steps of the signaling pathway the parasites were involved. Human THP-1 and mouse RAW264.7 macrophages were infected by SC10H2 promastigotes followed by cycloheximide stimulation to assess the alteration of intrinsic apoptosis in these cells. The results indicated that SC10H2 infection of human THP-1 macrophages could promote the initiation of intrinsic apoptosis, but completely opposite results were found in mouse RAW264.7 macrophages. Nevertheless, the expression of Bcl-2 and the DNA fragmentation rates were not altered by SC10H2 infection in the cell lines used in the experiments. This study suggests that SC10H2 promastigote infection is able to promote and delay the transduction of early apoptotic signals induced by cycloheximide in THP-1 and RAW264.7 macrophages, revealing that the regulation of intrinsic apoptosis in host cells by SC10H2 in vitro occurs in a host cell-dependent manner. The data from this study might play a significant role in further understanding the relationship between Leishmania and different host cells.
Collapse
|
225
|
Tranchemontagne ZR, Camire RB, O'Donnell VJ, Baugh J, Burkholder KM. Staphylococcus aureus Strain USA300 Perturbs Acquisition of Lysosomal Enzymes and Requires Phagosomal Acidification for Survival inside Macrophages. Infect Immun 2016; 84:241-53. [PMID: 26502911 PMCID: PMC4694005 DOI: 10.1128/iai.00704-15] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 10/22/2015] [Indexed: 02/08/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) causes invasive, drug-resistant skin and soft tissue infections. Reports that S. aureus bacteria survive inside macrophages suggest that the intramacrophage environment may be a niche for persistent infection; however, mechanisms by which the bacteria might evade macrophage phagosomal defenses are unclear. We examined the fate of the S. aureus-containing phagosome in THP-1 macrophages by evaluating bacterial intracellular survival and phagosomal acidification and maturation and by testing the impact of phagosomal conditions on bacterial viability. Multiple strains of S. aureus survived inside macrophages, and in studies using the MRSA USA300 clone, the USA300-containing phagosome acidified rapidly and acquired the late endosome and lysosome protein LAMP1. However, fewer phagosomes containing live USA300 bacteria than those containing dead bacteria associated with the lysosomal hydrolases cathepsin D and β-glucuronidase. Inhibiting lysosomal hydrolase activity had no impact on intracellular survival of USA300 or other S. aureus strains, suggesting that S. aureus perturbs acquisition of lysosomal enzymes. We examined the impact of acidification on S. aureus intramacrophage viability and found that inhibitors of phagosomal acidification significantly impaired USA300 intracellular survival. Inhibition of macrophage phagosomal acidification resulted in a 30-fold reduction in USA300 expression of the staphylococcal virulence regulator agr but had little effect on expression of sarA, saeR, or sigB. Bacterial exposure to acidic pH in vitro increased agr expression. Together, these results suggest that S. aureus survives inside macrophages by perturbing normal phagolysosome formation and that USA300 may sense phagosomal conditions and upregulate expression of a key virulence regulator that enables its intracellular survival.
Collapse
Affiliation(s)
| | - Ryan B Camire
- Department of Nursing, University of New England, Biddeford, Maine, USA
| | | | - Jessfor Baugh
- Department of Biology, University of New England, Biddeford, Maine, USA
| | | |
Collapse
|
226
|
Lisovsky A, Chamberlain MD, Wells LA, Sefton MV. Cell Interactions with Vascular Regenerative MAA-Based Materials in the Context of Wound Healing. Adv Healthc Mater 2015; 4:2375-87. [PMID: 26010569 DOI: 10.1002/adhm.201500192] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 04/13/2015] [Indexed: 12/19/2022]
Abstract
In diabetic patients the development of chronic non-healing wounds is a common complication. A methacrylic acid-based biomaterial is a vascular regenerative material that enhances diabetic healing without the use of cells or growth factors. The bioactive nature of this material is thought to be associated with its anionic charge or surface chemistry. Contact between the methacrylic acid-based biomaterial and tissue begins with protein (including complement) adsorption and is followed by interaction of the biomaterial with resident and infiltrating cells in the wound bed (e.g., macrophages and endothelial cells). This results in changes to their surface receptors to activate phosphorylation cascades that lead to differential activation of signalling pathways such as those involving osteopontin and sonic hedgehog. These changes modulate the phenotype of the cells in the wound bed, eventually improving vessel formation and wound healing. Understanding the molecular and cellular mechanisms will have broad implications for biomaterials, not just the methacrylic acid-based material, and will facilitate the advancement of regenerative biomaterials for diverse applications.
Collapse
Affiliation(s)
- Alexandra Lisovsky
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; M5S 3G9 Canada
| | | | - Laura Anne Wells
- Department of Chemical Engineering; Queen's University; K7L 3N6 Canada
| | - Michael Vivian Sefton
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; 164 College Street, Suite 407 Toronto Ontario M5S 3G9 Canada
| |
Collapse
|
227
|
Grootaert C, Kamiloglu S, Capanoglu E, Van Camp J. Cell Systems to Investigate the Impact of Polyphenols on Cardiovascular Health. Nutrients 2015; 7:9229-55. [PMID: 26569293 PMCID: PMC4663590 DOI: 10.3390/nu7115462] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 10/21/2015] [Accepted: 10/28/2015] [Indexed: 02/07/2023] Open
Abstract
Polyphenols are a diverse group of micronutrients from plant origin that may serve as antioxidants and that contribute to human health in general. More specifically, many research groups have investigated their protective effect against cardiovascular diseases in several animal studies and human trials. Yet, because of the excessive processing of the polyphenol structure by human cells and the residing intestinal microbial community, which results in a large variability between the test subjects, the exact mechanisms of their protective effects are still under investigation. To this end, simplified cell culture systems have been used to decrease the inter-individual variability in mechanistic studies. In this review, we will discuss the different cell culture models that have been used so far for polyphenol research in the context of cardiovascular diseases. We will also review the current trends in cell culture research, including co-culture methodologies. Finally, we will discuss the potential of these advanced models to screen for cardiovascular effects of the large pool of bioactive polyphenols present in foods and their metabolites.
Collapse
Affiliation(s)
- Charlotte Grootaert
- Laboratory of Food Chemistry and Human Nutrition, Department of Food Safety and Food Quality, Faculty of Bioscience Engineering, Ghent University, Coupure Links, Ghent 653 B-9000, Belgium.
| | - Senem Kamiloglu
- Laboratory of Food Chemistry and Human Nutrition, Department of Food Safety and Food Quality, Faculty of Bioscience Engineering, Ghent University, Coupure Links, Ghent 653 B-9000, Belgium.
- Department of Food Engineering, Faculty of Chemical and Metallurgical Engineering, Istanbul Technical University, Maslak 34469, Istanbul, Turkey.
| | - Esra Capanoglu
- Department of Food Engineering, Faculty of Chemical and Metallurgical Engineering, Istanbul Technical University, Maslak 34469, Istanbul, Turkey.
| | - John Van Camp
- Laboratory of Food Chemistry and Human Nutrition, Department of Food Safety and Food Quality, Faculty of Bioscience Engineering, Ghent University, Coupure Links, Ghent 653 B-9000, Belgium.
| |
Collapse
|
228
|
Zhang H, Qian PY, Ravasi T. Selective phosphorylation during early macrophage differentiation. Proteomics 2015; 15:3731-43. [PMID: 26307563 DOI: 10.1002/pmic.201400511] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 07/08/2015] [Accepted: 08/19/2015] [Indexed: 12/27/2022]
Abstract
The differentiation of macrophages from monocytes is a tightly controlled and complex biological process. Although numerous studies have been conducted using biochemical approaches or global gene/protein profiling, the mechanisms of the early stages of differentiation remain unclear. Here we used SILAC-based quantitative proteomics approach to perform temporal phosphoproteome profiling of early macrophage differentiation. We identified a large set of phosphoproteins and grouped them as PMA-regulated and non-regulated phosphoproteins in the early stages of differentiation. Further analysis of the PMA-regulated phosphoproteins revealed that transcriptional suppression, cytoskeletal reorganization and cell adhesion were among the most significantly activated pathways. Some key involved regulators of these pathways are mTOR, MYB, STAT1 and CTNNB. Moreover, we were able to classify the roles and activities of several transcriptional factors during different differentiation stages and found that E2F is likely to be an important regulator during the relatively late stages of differentiation. This study provides the first comprehensive picture of the dynamic phosphoproteome during myeloid cells differentiation, and identifies potential molecular targets in leukemic cells.
Collapse
Affiliation(s)
- Huoming Zhang
- Division of Biological and Environmental Sciences & Engineering, Division of Applied Mathematics and Computer Sciences, King Abdullah University of Science & Technology, Thuwal, Kingdom of Saudi Arabia.,Bioscience Core Laboratory, King Abdullah University of Science and Technology, Thuwal, Kingdom of Saudi Arabia
| | - Pei-Yuan Qian
- School of Science, Hong Kong University of Science and Technology, Hong Kong, P. R. China
| | - Timothy Ravasi
- Division of Biological and Environmental Sciences & Engineering, Division of Applied Mathematics and Computer Sciences, King Abdullah University of Science & Technology, Thuwal, Kingdom of Saudi Arabia
| |
Collapse
|
229
|
Kulkarni NN, Gunnarsson HI, Yi Z, Gudmundsdottir S, Sigurjonsson OE, Agerberth B, Gudmundsson GH. Glucocorticoid dexamethasone down-regulates basal and vitamin D3 induced cathelicidin expression in human monocytes and bronchial epithelial cell line. Immunobiology 2015; 221:245-52. [PMID: 26358366 DOI: 10.1016/j.imbio.2015.09.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/01/2015] [Indexed: 01/29/2023]
Abstract
Glucocorticoids (GCs) have been extensively used as the mainstream treatment for chronic inflammatory disorders. The persistent use of steroids in the past decades and the association with secondary infections warrants for detailed investigation into their effects on the innate immune system and the therapeutic outcome. In this study, we analyse the effect of GCs on antimicrobial polypeptide (AMP) expression. We hypothesize that GC related side effects, including secondary infections are a result of compromised innate immune responses. Here, we show that treatment with dexamethasone (Dex) inhibits basal mRNA expression of the following AMPs; human cathelicidin, human beta defensin 1, lysozyme and secretory leukocyte peptidase 1 in the THP-1 monocytic cell-line (THP-1 monocytes). Furthermore, pre-treatment with Dex inhibits vitamin D3 induced cathelicidin expression in THP-1 monocytes, primary monocytes and in the human bronchial epithelial cell line BCi NS 1.1. We also demonstrate that treatment with the glucocorticoid receptor (GR) inhibitor RU486 counteracts Dex mediated down-regulation of basal and vitamin D3 induced cathelicidin expression in THP-1 monocytes. Moreover, we confirmed the anti-inflammatory effect of Dex. Pre-treatment with Dex inhibits dsRNA mimic poly IC induction of the inflammatory chemokine IP10 (CXCL10) and cytokine IL1B mRNA expression in THP-1 monocytes. These results suggest that GCs inhibit innate immune responses, in addition to exerting beneficial anti-inflammatory effects.
Collapse
Affiliation(s)
- Nikhil Nitin Kulkarni
- Biomedical Center and Department of Life and Environmental Sciences, University of Iceland, Reykjavík, Iceland
| | - Hörður Ingi Gunnarsson
- Biomedical Center and Department of Life and Environmental Sciences, University of Iceland, Reykjavík, Iceland
| | - Zhiqian Yi
- Biomedical Center and Department of Life and Environmental Sciences, University of Iceland, Reykjavík, Iceland
| | - Steinunn Gudmundsdottir
- Faculty of Medicine, School of Health Sciences, Department of Anatomy, Biomedical Center, University of Iceland, Reykjavik, Iceland
| | - Olafur E Sigurjonsson
- Biomedical Center and Department of Life and Environmental Sciences, University of Iceland, Reykjavík, Iceland; The Blood Bank, Landspitali University Hospital, Reykjavik, Iceland; Institute of Biomedical and Neural Engineering, School of Science and Engineering, Reykjavik University, Reykjavik, Iceland
| | - Birgitta Agerberth
- Department of Laboratory Medicine, Division of Clinical Microbiology Karolinska University Hospital, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Gudmundur H Gudmundsson
- Biomedical Center and Department of Life and Environmental Sciences, University of Iceland, Reykjavík, Iceland.
| |
Collapse
|
230
|
Lauric acid abolishes interferon-gamma (IFN-γ)-induction of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) expression in human macrophages. ASIAN PACIFIC JOURNAL OF REPRODUCTION 2015. [DOI: 10.1016/j.apjr.2015.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
231
|
Luque MCA, Gutierrez PS, Debbas V, Kalil J, Stolf BS. CD100 and plexins B2 and B1 mediate monocyte-endothelial cell adhesion and might take part in atherogenesis. Mol Immunol 2015; 67:559-67. [PMID: 26275342 DOI: 10.1016/j.molimm.2015.07.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 06/23/2015] [Accepted: 07/22/2015] [Indexed: 02/01/2023]
Abstract
Leukocyte migration is essential for the function of the immune system. Their recruitment from the vessels to the tissues involves sequential molecular interactions between leukocytes and endothelial cells (ECs). Many adhesion molecules involved in this process have already been described. However, additional molecules may be important in this interaction, and here we explore the potential role for CD100 and plexins in monocyte-EC binding. CD100 was shown to be involved in platelet-endothelial cell interaction, an important step in atherogenesis and thrombus formation. In a recent work we have described CD100 expression in monocytes and in macrophages and foam cells of human atherosclerotic plaques. In the present work, we have identified plexin B2 as a putative CD100 receptor in these cells. We have detected CD100 expression in the endothelium as well as in in vitro cultured endothelial cells. Blocking of CD100, plexin B1 and/or B2 in adhesion experiments have shown that both CD100 and plexins act as adhesion molecules involved in monocyte-endothelial cell binding. This effect may be mediated by CD100 expressed in both cell types, probably coupled to the receptors endothelial plexin B1 and monocytic plexin B2. These results can bring new insights about a possible biological activity of CD100 in monocyte adhesion and atherosclerosis, as well as a future candidate for targeting therapeutics.
Collapse
Affiliation(s)
- Maria Carolina A Luque
- Heart Institute of São Paulo (InCor), HC-FMUSP, São Paulo, SP, Brazil; Clinical Immunology and Allergy, Department of Clinical Medicine, University of São Paulo Medical School-HC-FMUSP, São Paulo, SP, Brazil
| | - Paulo S Gutierrez
- Heart Institute of São Paulo (InCor), HC-FMUSP, São Paulo, SP, Brazil
| | - Victor Debbas
- Heart Institute of São Paulo (InCor), HC-FMUSP, São Paulo, SP, Brazil
| | - Jorge Kalil
- Heart Institute of São Paulo (InCor), HC-FMUSP, São Paulo, SP, Brazil; Clinical Immunology and Allergy, Department of Clinical Medicine, University of São Paulo Medical School-HC-FMUSP, São Paulo, SP, Brazil; Institute for Investigation in Immunology - INCT - National Institute of Science and Technology, São Paulo, SP, Brazil
| | - Beatriz S Stolf
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
232
|
Tarasova NK, Ytterberg AJ, Lundberg K, Zhang XM, Harris RA, Zubarev RA. Proteomics Reveals a Role for Attachment in Monocyte Differentiation into Efficient Proinflammatory Macrophages. J Proteome Res 2015. [PMID: 26216291 DOI: 10.1021/acs.jproteome.5b00659] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Monocytes are blood-borne cells of the innate immune system. They can be differentiated and activated into proinflammatory macrophages that might be employed in tumor immune therapy. Monocyte exposure to lipopolysaccharide (LPS) is a standard method to induce a proinflammatory macrophage state, with the resultant population comprising both adherent and nonadherent cells. In the current study, we aimed to identify the differences in proteomes of these monocyte subpopulations, which addresses a more general question about the role of attachment in monocyte differentiation. Label-free proteomics of a model of human monocytes (THP-1 cell line) revealed that the cells remaining in suspension upon LPS treatment were activated by cytokines and primed for rapid responsiveness to pathogens. In terms of proteome change, the adhesion process was orthogonal to activation. Adherent cells exhibited signs of differentiation and enhanced innate immune responsivity, being closer to macrophages. These findings indicate that adherent, LPS-treated cells would be more appropriate for use in tumor therapeutic applications.
Collapse
Affiliation(s)
| | | | - Karin Lundberg
- Centre for Molecular Medicine, Karolinska Hospital , SE 17176 Stockholm, Sweden
| | - Xing-Mei Zhang
- Centre for Molecular Medicine, Karolinska Hospital , SE 17176 Stockholm, Sweden
| | - Robert A Harris
- Centre for Molecular Medicine, Karolinska Hospital , SE 17176 Stockholm, Sweden
| | | |
Collapse
|
233
|
Huynh DTN, Kim AY, Seol IH, Jung S, Lim MC, Lee JA, Jo MR, Choi SJ, Kim B, Lee J, Kim W, Kim YR. Inactivation of the virulence factors from 2,3-butanediol-producing Klebsiella pneumoniae. Appl Microbiol Biotechnol 2015; 99:9427-38. [PMID: 26239074 DOI: 10.1007/s00253-015-6861-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 07/13/2015] [Accepted: 07/16/2015] [Indexed: 01/06/2023]
Abstract
The microbiological production of 2,3-butanediol (2,3-BDO) has attracted considerable attention as an alternative way to produce high-value chemicals from renewable sources. Among the number of 2,3-BDO-producing microorganisms, Klebsiella pneumoniae has been studied most extensively and is known to produce large quantity of 2,3-BDO from a range of substrates. On the other hand, the pathogenic characteristics of the bacteria have limited its industrial applications. In this study, two major virulence traits, outer core LPS and fimbriae, were removed through homologous recombination from 2,3-BDO-producing K. pneumoniae 2242 to expand its uses to the industrial scale. The K. pneumoniae 2242 ∆wabG mutant strain was found to have an impaired capsule, which significantly reduced its ability to bind to the mucous layer and evade the phagocytic activity of macrophage. The association with the human ileocecal epithelial cell, HCT-8, and the bladder epithelial cell, T-24, was also reduced dramatically in the K. pneumoniae 2242 ∆fimA mutant strain that was devoid of fimbriae. However, the growth rate and production yield for 2,3-BDO were unaffected. The K. pneumoniae strains developed in this study, which are devoid of the major virulence factors, have a high potential for the efficient and sustainable production of 2,3-BDO.
Collapse
Affiliation(s)
- Duyen Thi Ngoc Huynh
- Graduate School of Biotechnology, Kyung Hee University, Yongin, 446-701, Republic of Korea.,Department of Food Science and Biotechnology, Kyung Hee University, Yongin, 446-701, Republic of Korea
| | - Ah-Young Kim
- Graduate School of Biotechnology, Kyung Hee University, Yongin, 446-701, Republic of Korea.,Department of Food Science and Biotechnology, Kyung Hee University, Yongin, 446-701, Republic of Korea
| | - In-Hye Seol
- Graduate School of Biotechnology, Kyung Hee University, Yongin, 446-701, Republic of Korea.,Department of Food Science and Biotechnology, Kyung Hee University, Yongin, 446-701, Republic of Korea
| | - Samuel Jung
- Graduate School of Biotechnology, Kyung Hee University, Yongin, 446-701, Republic of Korea.,Department of Food Science and Biotechnology, Kyung Hee University, Yongin, 446-701, Republic of Korea
| | - Min-Cheol Lim
- Graduate School of Biotechnology, Kyung Hee University, Yongin, 446-701, Republic of Korea.,Department of Food Science and Biotechnology, Kyung Hee University, Yongin, 446-701, Republic of Korea
| | - Jeong-A Lee
- Department of Food Science and Technology, Seoul Women's University, Seoul, 139-774, Republic of Korea
| | - Mi-Rae Jo
- Department of Food Science and Technology, Seoul Women's University, Seoul, 139-774, Republic of Korea
| | - Soo-Jin Choi
- Department of Food Science and Technology, Seoul Women's University, Seoul, 139-774, Republic of Korea
| | - Borim Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 121-742, Republic of Korea
| | - Jinwon Lee
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 121-742, Republic of Korea
| | - Wooki Kim
- Graduate School of Biotechnology, Kyung Hee University, Yongin, 446-701, Republic of Korea.,Department of Food Science and Biotechnology, Kyung Hee University, Yongin, 446-701, Republic of Korea
| | - Young-Rok Kim
- Graduate School of Biotechnology, Kyung Hee University, Yongin, 446-701, Republic of Korea. .,Department of Food Science and Biotechnology, Kyung Hee University, Yongin, 446-701, Republic of Korea.
| |
Collapse
|
234
|
Anand N, Kanwar RK, Dubey ML, Vahishta RK, Sehgal R, Verma AK, Kanwar JR. Effect of lactoferrin protein on red blood cells and macrophages: mechanism of parasite-host interaction. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:3821-35. [PMID: 26251568 PMCID: PMC4524381 DOI: 10.2147/dddt.s77860] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Lactoferrin is a natural multifunctional protein known to have antitumor, antimicrobial, and anti-inflammatory activity. Apart from its antimicrobial effects, lactoferrin is known to boost the immune response by enhancing antioxidants. Lactoferrin exists in various forms depending on its iron saturation. The present study was done to observe the effect of lactoferrin, isolated from bovine and buffalo colostrum, on red blood cells (RBCs) and macrophages (human monocytic cell line-derived macrophages THP1 cells). Methods Lactoferrin obtained from both species and in different iron saturation forms were used in the present study, and treatment of host cells were given with different forms of lactoferrin at different concentrations. These treated host cells were used for various studies, including morphometric analysis, viability by MTT assay, survivin gene expression, production of reactive oxygen species, phagocytic properties, invasion assay, and Toll-like receptor-4, Toll-like receptor-9, and MDR1 expression, to investigate the interaction between lactoferrin and host cells and the possible mechanism of action with regard to parasitic infections. Results The mechanism of interaction between host cells and lactoferrin have shown various aspects of gene expression and cellular activity depending on the degree of iron saturation of lactoferrin. A significant increase (P<0.05) in production of reactive oxygen species, phagocytic activity, and Toll-like receptor expression was observed in host cells incubated with iron-saturated lactoferrin when compared with an untreated control group. However, there was no significant (P>0.05) change in percentage viability in the different groups of host cells treated, and no downregulation of survivin gene expression was found at 48 hours post-incubation. Upregulation of the Toll-like receptor and downregulation of the P-gp gene confirmed the immunomodulatory potential of lactoferrin protein. Conclusion The present study details the interaction between lactoferrin and parasite host cells, ie, RBCs and macrophages, using various cellular processes and expression studies. The study reveals the possible mechanism of action against various intracellular pathogens such as Toxoplasma, Plasmodium, Leishmania, Trypanosoma, and Mycobacterium. The presence of iron in lactoferrin plays an important role in enhancing the various activities taking place inside these cells. This work provides a lot of information about targeting lactoferrin against many parasitic infections which can rule out the exact pathways for inhibition of diseases caused by intracellular microbes mainly targeting RBCs and macrophages for their survival. Therefore, this initial study can serve as a baseline for further evaluation of the mechanism of action of lactoferrin against parasitic diseases, which is not fully understood to date.
Collapse
Affiliation(s)
- Namrata Anand
- Department of Medical Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rupinder K Kanwar
- Nanomedicine Laboratory of Immunology and Molecular Biomedical Research, School of Medicine, Molecular and Medical Research Strategic Research Centre, Faculty of Health, Deakin University, Geelong, VIC, Australia
| | - Mohan Lal Dubey
- Department of Medical Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - R K Vahishta
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh
| | - Rakesh Sehgal
- Department of Medical Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Anita K Verma
- Nanobiotech Laboratory, Department of Zoology, Kirorimal College, University of Delhi, Delhi, India
| | - Jagat R Kanwar
- Nanomedicine Laboratory of Immunology and Molecular Biomedical Research, School of Medicine, Molecular and Medical Research Strategic Research Centre, Faculty of Health, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
235
|
Cha SJ, Park K, Srinivasan P, Schindler CW, van Rooijen N, Stins M, Jacobs-Lorena M. CD68 acts as a major gateway for malaria sporozoite liver infection. ACTA ACUST UNITED AC 2015. [PMID: 26216124 PMCID: PMC4548058 DOI: 10.1084/jem.20110575] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Cha et al. use a phage display library screen to identify a peptide, P39, that binds to CD68 on the surface of Kupffer cells to inhibit malaria sporozoite cell entry. Thus, P39 may represent a therapeutic strategy for malaria by limiting hepatic infection. After being delivered by the bite from an infected mosquito, Plasmodium sporozoites enter the blood circulation and infect the liver. Previous evidence suggests that Kupffer cells, a macrophage-like component of the liver blood vessel lining, are traversed by sporozoites to initiate liver invasion. However, the molecular determinants of sporozoite–Kupffer cell interactions are unknown. Understanding the molecular basis for this specific recognition may lead to novel therapeutic strategies to control malaria. Using a phage display library screen, we identified a peptide, P39, that strongly binds to the Kupffer cell surface and, importantly, inhibits sporozoite Kupffer cell entry. Furthermore, we determined that P39 binds to CD68, a putative receptor for sporozoite invasion of Kupffer cells that acts as a gateway for malaria infection of the liver.
Collapse
Affiliation(s)
- Sung-Jae Cha
- W. Harry Feinstone Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health; and Department of Neurology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21205 W. Harry Feinstone Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health; and Department of Neurology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21205
| | - Kiwon Park
- W. Harry Feinstone Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health; and Department of Neurology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21205 W. Harry Feinstone Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health; and Department of Neurology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21205
| | - Prakash Srinivasan
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | - Christian W Schindler
- Department of Microbiology and Immunology and Department of Medicine, Columbia University, New York, NY 10032 Department of Microbiology and Immunology and Department of Medicine, Columbia University, New York, NY 10032
| | - Nico van Rooijen
- Department of Molecular Cell Biology and Immunology, VUmc, 1081 BT Amsterdam, Netherlands
| | - Monique Stins
- W. Harry Feinstone Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health; and Department of Neurology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21205
| | - Marcelo Jacobs-Lorena
- W. Harry Feinstone Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health; and Department of Neurology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21205 W. Harry Feinstone Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health; and Department of Neurology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21205
| |
Collapse
|
236
|
Couleau N, Falla J, Beillerot A, Battaglia E, D’Innocenzo M, Plançon S, Laval-Gilly P, Bennasroune A. Effects of Endocrine Disruptor Compounds, Alone or in Combination, on Human Macrophage-Like THP-1 Cell Response. PLoS One 2015; 10:e0131428. [PMID: 26133781 PMCID: PMC4489735 DOI: 10.1371/journal.pone.0131428] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 06/01/2015] [Indexed: 12/21/2022] Open
Abstract
The aim of the present study was to evaluate the immunological effects on human macrophages of four endocrine disruptor compounds (EDCs) using the differentiated human THP-1 cell line as a model. We studied first the effects of these EDCs, including Bisphenol A (BPA), di-ethylhexyl-phthalate (DEHP), dibutyl phthalate (DBP) and 4-tert-octylphenol (4-OP), either alone or in combination, on cytokine secretion, and phagocytosis. We then determined whether or not these effects were mediated by estrogen receptors via MAPK pathways. It was found that all four EDCs studied reduced strongly the phagocytosis of the differentiated THP-1 cells and that several of these EDCs disturbed also TNF-α, IL-1 β and IL-8 cytokine secretions. Furthermore, relative to control treatment, decreased ERK 1/2 phosphorylation was always associated with EDCs treatments-either alone or in certain combinations (at 0.1 μM for each condition). Lastly, as treatments by an estrogen receptor antagonist suppressed the negative effects on ERK 1/2 phosphorylation observed in cells treated either alone with BPA, DEHP, 4-OP or with the combined treatment of BPA and DEHP, we suggested that estrogen receptor-dependent pathway is involved in mediating the effects of EDCs on human immune system. Altogether, these results advocate that EDCs can disturb human immune response at very low concentrations.
Collapse
Affiliation(s)
- N. Couleau
- Université de Lorraine, CNRS UMR 7360, Laboratoire Interdisciplinaire des Environnements Continentaux (LIEC), IUT Thionville-Yutz, Espace Cormontaigne, Yutz, France
| | - J. Falla
- Université de Lorraine, CNRS UMR 7360, Laboratoire Interdisciplinaire des Environnements Continentaux (LIEC), IUT Thionville-Yutz, Espace Cormontaigne, Yutz, France
| | - A. Beillerot
- IUT Thionville-Yutz, Impasse Alfred Kastler Espace Cormontaigne, Yutz, France
| | - E. Battaglia
- Université de Lorraine, CNRS UMR 7360, Laboratoire Interdisciplinaire des Environnements Continentaux (LIEC), Campus Bridoux—8, Metz, France
| | - M. D’Innocenzo
- IUT Thionville-Yutz, Impasse Alfred Kastler Espace Cormontaigne, Yutz, France
| | - S. Plançon
- Calcium Signaling and Inflammation Group, Life Sciences Research Unit, University of Luxembourg, Luxembourg, Luxembourg
| | - P. Laval-Gilly
- Université de Lorraine, CNRS UMR 7360, Laboratoire Interdisciplinaire des Environnements Continentaux (LIEC), IUT Thionville-Yutz, Espace Cormontaigne, Yutz, France
| | - A. Bennasroune
- Université de Lorraine, CNRS UMR 7360, Laboratoire Interdisciplinaire des Environnements Continentaux (LIEC), IUT Thionville-Yutz, Espace Cormontaigne, Yutz, France
| |
Collapse
|
237
|
Gutowska I, Baranowska-Bosiacka I, Goschorska M, Kolasa A, Łukomska A, Jakubczyk K, Dec K, Chlubek D. Fluoride as a factor initiating and potentiating inflammation in THP1 differentiated monocytes/macrophages. Toxicol In Vitro 2015; 29:1661-8. [PMID: 26119525 DOI: 10.1016/j.tiv.2015.06.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 06/23/2015] [Accepted: 06/25/2015] [Indexed: 02/08/2023]
Abstract
It is well known that exposure to fluorides lead to an increased ROS production and enhances the inflammatory reactions. Therefore we decided to examine whether cyclooxygenases (particular COX-2) activity and expression may be changed by fluoride in THP1 macrophages and in this way may change the prostanoids biosynthesis. In the present work we demonstrate that fluoride increased concentration of PGE2 and TXA2 in THP1 macrophages. Following exposure to 1-10 μM NaF, COX-2 protein and COX-2 transcript increased markedly. COX-2 protein up-regulation probably is mediated by ROS, produced during fluoride-induced inflammatory reactions. Additional fluoride activates the transcription factor, nuclear factor (NF)-kappaB, which is involved in the up-regulation of COX-2 gene expression. This study indicated that even in small concentrations fluoride changes the amounts and activity of COX-1 and COX-2 enzymes taking part in the initiating and development of inflammatory process.
Collapse
Affiliation(s)
- I Gutowska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University, Broniewskiego 24 Str., Szczecin, Poland
| | - I Baranowska-Bosiacka
- Department of Biochemistry, Pomeranian Medical University, Powstańców Wlkp 72 Str., Szczecin, Poland.
| | - M Goschorska
- Department of Biochemistry, Pomeranian Medical University, Powstańców Wlkp 72 Str., Szczecin, Poland
| | - A Kolasa
- Department of Histology and Embryology, Pomeranian Medical University, Powstańców Wlkp 72 Str., Szczecin, Poland
| | - A Łukomska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University, Broniewskiego 24 Str., Szczecin, Poland
| | - K Jakubczyk
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University, Broniewskiego 24 Str., Szczecin, Poland
| | - K Dec
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University, Broniewskiego 24 Str., Szczecin, Poland
| | - D Chlubek
- Department of Biochemistry, Pomeranian Medical University, Powstańców Wlkp 72 Str., Szczecin, Poland
| |
Collapse
|
238
|
Ankathatti Munegowda M, Hu J. Transient blocking of NK cell function with small molecule inhibitors for helper dependant adenoviral vector-mediated gene delivery. Cell Biosci 2015; 5:29. [PMID: 26085921 PMCID: PMC4470062 DOI: 10.1186/s13578-015-0023-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 06/05/2015] [Indexed: 11/10/2022] Open
Abstract
One major challenge in gene therapy is the host immune responses against viral vectors. Previous studies indicate the involvement of NK cells in stunted gene expression in viral vector mediated gene therapy. To understand the problem of the immune responses, we have developed an in-vitro co-culture system with human NK cell line, macrophages and airway epithelial cells. We showed that small molecule blockers, CAPE and ruxolitinib, for NF-κB and JAK-STAT pathways, respectively, significantly inhibited cytokine secretion by macrophages. When NK cells are co-cultured with helper-dependent adenoviral (HD-Ad) vector activated macrophages, IFN-γ cytokine expression by NK cells increased significantly, which was inhibited effectively by ruxolitinib and CAPE, and there was an additive effect when both inhibitors were used. We demonstrated that NK cells activated by cytokines produced by HD-Ad-activated macrophages kill HD-Ad vector transduced bronchial epithelial cells. This cell killing activity was significantly reduced by CAPE and ruxolitinib. Combination of these two inhibitors had an additive effect on inhibiting NK cell mediate killing of gene transduced cells. Transient inhibition of NK cell response at its peak may enhance sustained gene expression. Our data suggest that combination of CAPE and ruxolitinib may help in protecting gene transduced airway epithelial cells to prolong transgene expression.
Collapse
Affiliation(s)
- Manjunatha Ankathatti Munegowda
- Department of Physiology & Experimental Medicine, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning (PGCRL), 9th floor, 686 Bay Street, Toronto, ON M5G 0A4 Canada ; University of Toronto, Toronto, ON Canada
| | - Jim Hu
- Department of Physiology & Experimental Medicine, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning (PGCRL), 9th floor, 686 Bay Street, Toronto, ON M5G 0A4 Canada ; University of Toronto, Toronto, ON Canada
| |
Collapse
|
239
|
Ward R, Sims AH, Lee A, Lo C, Wynne L, Yusuf H, Gregson H, Lisanti MP, Sotgia F, Landberg G, Lamb R. Monocytes and macrophages, implications for breast cancer migration and stem cell-like activity and treatment. Oncotarget 2015; 6:14687-99. [PMID: 26008983 PMCID: PMC4546497 DOI: 10.18632/oncotarget.4189] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 04/08/2015] [Indexed: 12/17/2022] Open
Abstract
Macrophages are a major cellular constituent of the tumour stroma and contribute to breast cancer prognosis. The precise role and treatment strategies to target macrophages remain elusive. As macrophage infiltration is associated with poor prognosis and high grade tumours we used the THP-1 cell line to model monocyte-macrophage differentiation in co-culture with four breast cancer cell lines (MCF7, T47D, MDA-MB-231, MDA-MB-468) to model in vivo cellular interactions. Polarisation into M1 and M2 subtypes was confirmed by specific cell marker expression of ROS and HLA-DR, respectively. Co-culture with all types of macrophage increased migration of ER-positive breast cancer cell lines, while M2-macrophages increased mammosphere formation, compared to M1-macrophages, in all breast cancer cells lines. Treatment of cells with Zoledronate in co-culture reduced the "pro-tumourigenic" effects (increased mammospheres/migration) exerted by macrophages. Direct treatment of breast cancer cells in homotypic culture was unable to reduce migration or mammosphere formation.Macrophages promote "pro-tumourigenic" cellular characteristics of breast cancer cell migration and stem cell activity. Zoledronate targets macrophages within the microenvironment which in turn, reduces the "pro-tumourigenic" characteristics of breast cancer cells. Zoledronate offers an exciting new treatment strategy for both primary and metastatic breast cancer.
Collapse
Affiliation(s)
- Rebecca Ward
- Breakthrough Breast Cancer Unit, Institute of Cancer Sciences, Paterson Institute for Cancer Research, University of Manchester, Manchester, UK
| | - Andrew H. Sims
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research Centre, Carrington Crescent, Edinburgh, UK
| | - Alexander Lee
- Breakthrough Breast Cancer Unit, Institute of Cancer Sciences, Paterson Institute for Cancer Research, University of Manchester, Manchester, UK
| | - Christina Lo
- Breakthrough Breast Cancer Unit, Institute of Cancer Sciences, Paterson Institute for Cancer Research, University of Manchester, Manchester, UK
| | - Luke Wynne
- Breakthrough Breast Cancer Unit, Institute of Cancer Sciences, Paterson Institute for Cancer Research, University of Manchester, Manchester, UK
| | - Humza Yusuf
- Breakthrough Breast Cancer Unit, Institute of Cancer Sciences, Paterson Institute for Cancer Research, University of Manchester, Manchester, UK
| | - Hannah Gregson
- Breakthrough Breast Cancer Unit, Institute of Cancer Sciences, Paterson Institute for Cancer Research, University of Manchester, Manchester, UK
| | - Michael P. Lisanti
- Breakthrough Breast Cancer Unit, Institute of Cancer Sciences, Paterson Institute for Cancer Research, University of Manchester, Manchester, UK
| | - Federica Sotgia
- Breakthrough Breast Cancer Unit, Institute of Cancer Sciences, Paterson Institute for Cancer Research, University of Manchester, Manchester, UK
| | - Göran Landberg
- Breakthrough Breast Cancer Unit, Institute of Cancer Sciences, Paterson Institute for Cancer Research, University of Manchester, Manchester, UK
- Sahlgrenska Cancer Center, University of Gothenburg, Sweden
| | - Rebecca Lamb
- Breakthrough Breast Cancer Unit, Institute of Cancer Sciences, Paterson Institute for Cancer Research, University of Manchester, Manchester, UK
| |
Collapse
|
240
|
Chiu KC, Lee CH, Liu SY, Yeh CT, Huang RY, Yuh DY, Cheng JC, Chou YT, Shieh YS. Protumoral effect of macrophage through Axl activation on mucoepidermoid carcinoma. J Oral Pathol Med 2015; 43:538-44. [PMID: 25184164 DOI: 10.1111/jop.12163] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This study aims to test the potential involvement of Axl signaling in the protumoral effect of tumor-associated macrophages (TAMs) in mucoepidermoid carcinoma (MEC). MATERIALS AND METHODS We carried out cocultured experiments by incubation of MEC cells (UTMUC-1) and macrophages (THP-1) and examined Axl activation status. The expression of MMPs and behavior change were examined in UT-MUC-1 cells. The effect of Axl signaling on co-cultured cancer cells was further investigated by knockdown Axl expression and suppression by Axl-specific inhibitor R428. RESULTS Activation of Axl signaling and increased expression and activity of MMP-2 and MMP-9 along with increased invasion/migration ability in MEC cells were observed when co-cultured with TAMs. Upon knockdown of Axl in MEC or addition of R428 in the co-cultured system, these co-cultured effects were diminished. CONCLUSION TAMs play a protumoral role in MEC via activation of the Axl signaling pathway, up-regulating MMPs expression, and increasing invasion/migration ability.
Collapse
|
241
|
de Vallière C, Wang Y, Eloranta JJ, Vidal S, Clay I, Spalinger MR, Tcymbarevich I, Terhalle A, Ludwig MG, Suply T, Fried M, Kullak-Ublick GA, Frey-Wagner I, Scharl M, Seuwen K, Wagner CA, Rogler G. G Protein-coupled pH-sensing Receptor OGR1 Is a Regulator of Intestinal Inflammation. Inflamm Bowel Dis 2015; 21:1269-81. [PMID: 25856770 PMCID: PMC4450952 DOI: 10.1097/mib.0000000000000375] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 01/27/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND A novel family of proton-sensing G protein-coupled receptors, including OGR1, GPR4, and TDAG8, was identified to be important for physiological pH homeostasis and inflammation. Thus, we determined the function of proton-sensing OGR1 in the intestinal mucosa. MTEHODS OGR1 expression in colonic tissues was investigated in controls and patients with IBD. Expression of OGR1 upon cell activation was studied in the Mono Mac 6 (MM6) cell line and primary human and murine monocytes by real-time PCR. Ogr1 knockout mice were crossbred with Il-10 deficient mice and studied for more than 200 days. Microarray profiling was performed using Ogr1 and Ogr1 (WT) residential peritoneal macrophages. RESULTS Patients with IBD expressed higher levels of OGR1 in the mucosa than non-IBD controls. Treatment of MM6 cells with TNF, led to significant upregulation of OGR1 expression, which could be reversed by the presence of NF-κB inhibitors. Kaplan-Meier survival analysis showed a significantly delayed onset and progression of rectal prolapse in female Ogr1/Il-10 mice. These mice displayed significantly less rectal prolapses. Upregulation of gene expression, mediated by OGR1, in response to extracellular acidification in mouse macrophages was enriched for inflammation and immune response, actin cytoskeleton, and cell-adhesion gene pathways. CONCLUSIONS OGR1 expression is induced in cells of human macrophage lineage and primary human monocytes by TNF. NF-κB inhibition reverses the induction of OGR1 expression by TNF. OGR1 deficiency protects from spontaneous inflammation in the Il-10 knockout model. Our data indicate a pathophysiological role for pH-sensing receptor OGR1 during the pathogenesis of mucosal inflammation.
Collapse
Affiliation(s)
- Cheryl de Vallière
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
| | - Yu Wang
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
- Institute of Physiology, University of Zürich, Zürich, Switzerland
| | - Jyrki J. Eloranta
- Department of Clinical Pharmacology and Toxicology, University Hospital Zürich, Zürich, Switzerland; and
| | - Solange Vidal
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Ieuan Clay
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Marianne R. Spalinger
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
| | - Irina Tcymbarevich
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
| | - Anne Terhalle
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
| | | | - Thomas Suply
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Michael Fried
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
| | - Gerd A. Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital Zürich, Zürich, Switzerland; and
| | - Isabelle Frey-Wagner
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
| | - Michael Scharl
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
| | - Klaus Seuwen
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | - Gerhard Rogler
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
242
|
L-cystathionine inhibits oxidized low density lipoprotein-induced THP-1-derived macrophage inflammatory cytokine monocyte chemoattractant protein-1 generation via the NF-κB pathway. Sci Rep 2015; 5:10453. [PMID: 26020416 PMCID: PMC4447071 DOI: 10.1038/srep10453] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 04/14/2015] [Indexed: 11/29/2022] Open
Abstract
This study aimed to explore whether and how L-cystathionine had any regulatory effect on the inflammatory response in THP-1-derived macrophages cultured in vitro under oxidized low-density lipoprotein (ox-LDL) stimulation. The human monocyte line THP-1 cell was cultured in vitro and differentiated into macrophages after 24 hours of PMA induction. Macrophages were pretreated with L-cystathionine and then treated with ox-LDL. The results showed that compared with the controls, ox-LDL stimulation significantly upregulated the expression of THP-1-derived macrophage MCP-1 by enhancing NF-κB p65 phosphorylation, nuclear translocation and DNA binding with the MCP-1 promoter. Compared with the ox-LDL group, 0.3 mmol/L and 1.0 mmol/L L-cystathionine significantly inhibited the expression of THP-1-derived macrophage MCP-1. Mechanistically, 0.3 mmol/L and 1.0 mmol/L L-cystathionine suppressed phosphorylation and nuclear translocation of the NF-κB p65 protein, as well as the DNA binding activity and DNA binding level of NF-κB with the MCP-1 promoter, which resulted in a reduced THP-1-derived macrophage MCP-1 generation. This study suggests that L-cystathionine could inhibit the expression of MCP-1 in THP-1-derived macrophages induced by ox-LDL via inhibition of NF-κB p65 phosphorylation, nuclear translocation, and binding of the MCP-1 promoter sequence after entry into the nucleus.
Collapse
|
243
|
Cheong WC, Kang HR, Yoon H, Kang SJ, Ting JPY, Song MJ. Influenza A Virus NS1 Protein Inhibits the NLRP3 Inflammasome. PLoS One 2015; 10:e0126456. [PMID: 25978411 PMCID: PMC4433236 DOI: 10.1371/journal.pone.0126456] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 04/02/2015] [Indexed: 12/23/2022] Open
Abstract
The inflammasome is a molecular platform that stimulates the activation of caspase-1 and the processing of pro-interleukin (IL)-1β and pro-IL-18 for secretion. The NOD-like receptor family, pyrin domain containing 3 (NLRP3) protein is activated by diverse molecules and pathogens, leading to the formation of the NLRP3 inflammasome. Recent studies showed that the NLRP3 inflammasome mediates innate immunity against influenza A virus (IAV) infection. In this study, we investigated the function of the IAV non-structural protein 1 (NS1) in the modulation of NLRP3 inflammasome. We found that NS1 proteins derived from both highly pathogenic and low pathogenic strains efficiently decreased secretion of IL-1β and IL-18 from THP-1 cells treated with LPS and ATP. NS1 overexpression significantly impaired the transcription of proinflammatory cytokines by inhibiting transactivation of the nuclear factor-κB (NF-κB), a major transcription activator. Furthermore, NS1 physically interacted with endogenous NLRP3 and activation of the NLRP3 inflammasome was abrogated in NS1-expressing THP-1 cells. These findings suggest that NS1 downregulates NLRP3 inflammasome activation by targeting NLRP3 as well as NF-κB, leading to a reduction in the levels of inflammatory cytokines as a viral immune evasion strategy.
Collapse
Affiliation(s)
- Woo-Chang Cheong
- Virus-Host Interactions Laboratory, Department of Biosystems and Biotechnology, Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 136–713, Republic of Korea
| | - Hye-Ri Kang
- Virus-Host Interactions Laboratory, Department of Biosystems and Biotechnology, Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 136–713, Republic of Korea
| | - Hyunyee Yoon
- Virus-Host Interactions Laboratory, Department of Biosystems and Biotechnology, Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 136–713, Republic of Korea
- Laboratory of Protein Immunology, Biomedical Research Institute, Seoul National University Hospital, Seoul, 110–744, Republic of Korea
| | - Suk-Jo Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 305–701, Republic of Korea
| | - Jenny P.-Y. Ting
- Departments of Genetics, Microbiology and Immunology, Center for Translational Immunology and Inflammatory Disease Institute, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, United States of America
| | - Moon Jung Song
- Virus-Host Interactions Laboratory, Department of Biosystems and Biotechnology, Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 136–713, Republic of Korea
- * E-mail:
| |
Collapse
|
244
|
Hamelin-Morrissette J, Cloutier S, Girouard J, Belgorosky D, Eiján AM, Legault J, Reyes-Moreno C, Bérubé G. Identification of an anti-inflammatory derivative with anti-cancer potential: The impact of each of its structural components on inflammatory responses in macrophages and bladder cancer cells. Eur J Med Chem 2015; 96:259-68. [DOI: 10.1016/j.ejmech.2015.04.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 04/09/2015] [Accepted: 04/10/2015] [Indexed: 11/26/2022]
|
245
|
Rudisch A, Dewhurst MR, Horga LG, Kramer N, Harrer N, Dong M, van der Kuip H, Wernitznig A, Bernthaler A, Dolznig H, Sommergruber W. High EMT Signature Score of Invasive Non-Small Cell Lung Cancer (NSCLC) Cells Correlates with NFκB Driven Colony-Stimulating Factor 2 (CSF2/GM-CSF) Secretion by Neighboring Stromal Fibroblasts. PLoS One 2015; 10:e0124283. [PMID: 25919140 PMCID: PMC4412534 DOI: 10.1371/journal.pone.0124283] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 03/12/2015] [Indexed: 12/23/2022] Open
Abstract
We established co-cultures of invasive or non-invasive NSCLC cell lines and various types of fibroblasts (FBs) to more precisely characterize the molecular mechanism of tumor-stroma crosstalk in lung cancer. The HGF-MET-ERK1/2-CREB-axis was shown to contribute to the onset of the invasive phenotype of Calu-1 with HGF being secreted by FBs. Differential expression analysis of the respective mono- and co-cultures revealed an upregulation of NFκB-related genes exclusively in co-cultures with Calu-1. Cytokine Array- and ELISA-based characterization of the “cytokine fingerprints” identified CSF2 (GM-CSF), CXCL1, CXCL6, VEGF, IL6, RANTES and IL8 as being specifically upregulated in various co-cultures. Whilst CXCL6 exhibited a strictly FB-type-specific induction profile regardless of the invasiveness of the tumor cell line, CSF2 was only induced in co-cultures of invasive cell lines regardless of the partnered FB type. These cultures revealed a clear link between the induction of CSF2 and the EMT signature of the cancer cell line. The canonical NFκB signaling in FBs, but not in tumor cells, was shown to be responsible for the induced and constitutive CSF2 expression. In addition to CSF2, cytokine IL6, IL8 and IL1B, and chemokine CXCL1 and CXCL6 transcripts were also shown to be increased in co-cultured FBs. In contrast, their induction was not strictly dependent on the invasiveness of the co-cultured tumor cell. In a multi-reporter assay, additional signaling pathways (AP-1, HIF1-α, KLF4, SP-1 and ELK-1) were found to be induced in FBs co-cultured with Calu-1. Most importantly, no difference was observed in the level of inducibility of these six signaling pathways with regard to the type of FBs used. Finally, upon tumor fibroblast interaction the massive induction of chemokines such as CXCL1 and CXCL6 in FBs might be responsible for increased recruitment of a monocytic cell line (THP-1) in a transwell assay.
Collapse
MESH Headings
- Animals
- Cadherins/metabolism
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Communication/drug effects
- Cell Line, Tumor
- Cytokines/metabolism
- Dermis/pathology
- Epithelial-Mesenchymal Transition/drug effects
- Epithelial-Mesenchymal Transition/genetics
- Fibroblasts/drug effects
- Fibroblasts/metabolism
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Regulatory Networks/drug effects
- Genes, Reporter
- Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
- Hepatocyte Growth Factor/pharmacology
- Humans
- Inflammation/pathology
- Kruppel-Like Factor 4
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- NF-kappa B/metabolism
- Neoplasm Invasiveness
- Phenotype
- Proto-Oncogene Proteins c-met/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Spheroids, Cellular/drug effects
- Spheroids, Cellular/metabolism
- Spheroids, Cellular/pathology
- Stromal Cells/metabolism
- Up-Regulation/drug effects
- Up-Regulation/genetics
Collapse
Affiliation(s)
- Albin Rudisch
- Department of Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
- Department of Microbiology, Immunobiology and Genetics, Center of Molecular Biology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | | | | | - Nina Kramer
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Nathalie Harrer
- Department of Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Meng Dong
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, Stuttgart, Baden-Württemberg, Germany
| | - Heiko van der Kuip
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, Stuttgart, Baden-Württemberg, Germany
| | - Andreas Wernitznig
- Department of Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Andreas Bernthaler
- Department of Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Helmut Dolznig
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Sommergruber
- Department of Lead Discovery, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
- * E-mail:
| |
Collapse
|
246
|
Lu J, Webster TJ. Reduced immune cell responses on nano and submicron rough titanium. Acta Biomater 2015; 16:223-31. [PMID: 25660564 DOI: 10.1016/j.actbio.2015.01.036] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 01/20/2015] [Accepted: 01/26/2015] [Indexed: 11/30/2022]
Abstract
Current bare metal stents can be improved by nanotechnology to support the simultaneous acceleration of endothelialization and consequent reduction of immune cell responses after implantation. In our prior study, electron beam deposition was utilized to create different scales of roughness on titanium stents including flat (F-Ti), a mixture of nanometer and submicron (S-Ti), and nanometer (N-Ti). Enhanced endothelial responses (adhesion, migration, and nitric acid/endothelin-1 secretion) on nanometer to submicron rough titanium were observed compared to flat titanium. The present study aimed to further investigate the influence of nano and submicron titanium surface features on immune cells. Initial monocyte adhesion was found to be reduced on nano and submicron surface features compared to a flat surface. In a model including both endothelial cells and monocytes, it was proven that the submicron surface gave rise to an endothelial cell monolayer which generated the highest amount of NOx and subsequently led to decreased adhesiveness of endothelial cells to monocytes. The analysis of monocyte morphology gave hints to less differentiated monocytes on a submicron surface. Furthermore, the adhesion of and pro-inflammatory cytokine release from macrophages were all reduced on nano and submicron titanium surface features compared to a flat surface. This study, thus, suggests that nano and submicron titanium surfaces should be further studied for improved vascular stent performance.
Collapse
Affiliation(s)
- Jing Lu
- School of Engineering, Brown University, Providence, RI 02912, USA
| | - Thomas J Webster
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA; Center of Excellence for Advanced Materials Research, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
247
|
Elsaidi HRH, Lowary TL. Effect of phenolic glycolipids from Mycobacterium kansasii on proinflammatory cytokine release. A structure-activity relationship study. Chem Sci 2015; 6:3161-3172. [PMID: 28706688 PMCID: PMC5490424 DOI: 10.1039/c4sc04004j] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Accepted: 03/25/2015] [Indexed: 12/13/2022] Open
Abstract
The cell wall of pathogenic mycobacteria is abundant with virulence factors, among which phenolic glycolipids (PGLs) are prominent examples. Mycobacterium kansasii, an important opportunistic pathogen, produces seven PGLs and their effect on the release of important proinflammatory cytokines that mediate disease progression has not been investigated. We previously showed that proinflammatory cytokines are modulated by PGLs from M. tuberculosis, M. leprae and M. bovis. In this paper we describe the synthesis of a series of 17 analogs of M. kansasii PGLs containing a truncated aglycone. Subsequently, the effect of these compounds on the release of proinflammatory cytokines (TNF-α, IL-6, IL-1β, MCP-1) and nitric oxide (NO) was evaluated. These compounds exerted an immunoinhibitory effect on the release of the tested cytokines. The concentration-dependent inhibitory profile of the tested molecules was also found to be dependent on the methylation pattern of the molecule and was mediated via toll-like receptor (TLR)-2. This study led to the discovery of a glycolipid (18) that shows promising potent anti-inflammatory properties making it a potential candidate for further optimization of its anti-inflammatory profile.
Collapse
Affiliation(s)
- Hassan R H Elsaidi
- Alberta Glycomics Centre and Department of Chemistry , University of Alberta , Edmonton , AB , Canada T6G 2G2 .
| | - Todd L Lowary
- Alberta Glycomics Centre and Department of Chemistry , University of Alberta , Edmonton , AB , Canada T6G 2G2 .
| |
Collapse
|
248
|
Michael DR, Davies TS, Laubertová L, Gallagher H, Ramji DP. The phosphoinositide 3-kinase signaling pathway is involved in the control of modified low-density lipoprotein uptake by human macrophages. Lipids 2015; 50:253-60. [PMID: 25663263 PMCID: PMC4339697 DOI: 10.1007/s11745-015-3993-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 01/19/2015] [Indexed: 02/05/2023]
Abstract
The transformation of macrophages into lipid-loaded foam cells is a critical early event in the pathogenesis of atherosclerosis. Both receptor-mediated uptake of modified LDL, mediated primarily by scavenger receptors-A (SR-A) and CD36 along with other proteins such as lipoprotein lipase (LPL), and macropinocytosis contribute to macrophage foam cell formation. The signaling pathways that are involved in the control of foam cell formation are not fully understood. In this study, we have investigated the role of phosphoinositide 3-kinase (PI3K) in relation to foam cell formation in human macrophages. The pan PI3K inhibitor LY294002 attenuated the uptake of modified LDL and macropinocytosis, as measured by Lucifer Yellow uptake, by human macrophages. In addition, the expression of SR-A, CD36 and LPL was attenuated by LY294002. The use of isoform-selective PI3K inhibitors showed that PI3K-β, -γ and -δ were all required for the expression of SR-A and CD36 whereas only PI3K-γ was necessary in the case of LPL. These studies reveal a pivotal role of PI3K in the control of macrophage foam cell formation and provide further evidence for their potential as therapeutic target against atherosclerosis.
Collapse
Affiliation(s)
- Daryn R. Michael
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX UK
| | - Thomas S. Davies
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX UK
| | - Lucia Laubertová
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX UK
- Institute of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University, Malá Hora 4, 036 01 Martin, Slovakia
| | - Hayley Gallagher
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX UK
| | - Dipak P. Ramji
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX UK
| |
Collapse
|
249
|
Ziaei A, Hoppstädter J, Kiemer AK, Ramezani M, Amirghofran Z, Diesel B. Inhibitory effects of teuclatriol, a sesquiterpene from salvia mirzayanii, on nuclear factor-κB activation and expression of inflammatory mediators. JOURNAL OF ETHNOPHARMACOLOGY 2015; 160:94-100. [PMID: 25446581 DOI: 10.1016/j.jep.2014.10.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 10/10/2014] [Accepted: 10/21/2014] [Indexed: 05/21/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Salvia mirzayanii Rech. f. & Esfand. is an endemic plant, which is only distributed in the south of Iran. In traditional Iranian medicine, the aerial parts of Salvia mirzayanii have been used for infections, inflammatory diseases, and as a tonic. From this plant, the sesquiterpene teuclatriol was isolated by bioactivity-guided fractionation due to its anti-proliferative actions on human lymphocytes. The guaiane sesquiterpene is lacking the methylene-γ-lactone function that is typically involved in the inhibiting properties of sesquiterpenes on NF-κB, a pivotal transcription factor in inflammatory processes. We here investigated anti-inflammatory effects of teuclatriol on human macrophage-like and endothelial cells. MATERIALS AND METHODS Non-toxic doses of teuclatriol were determined for both cell types by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide)-assay. The effect of teuclatriol on the activity of NF-κB in LPS-stimulated human monocytic THP-1 cells was studied using infrared electrophoretic mobility shift assay (IR-EMSA) using curcumin as positive control (32µM). THP-1 were differentiated into macrophage-like cells and evaluated for TNF-α secretion by L929 bioassay following stimulation with LPS and treatment with teuclatriol. Inflammatory gene expression in human umbilical vein endothelial cells (HUVEC), modeling target cells for TNF-α-induced inflammatory gene activation, was investigated by real-time RT-PCR. RESULTS The LPS-induced DNA binding activity of NF-κB in THP-1 was significantly decreased by non-toxic doses of teuclatriol (312 and 390µM). Teuclatriol reduced the production of TNF-α in a dose-dependent manner. mRNA levels of both monocyte chemoattractant protein (MCP)-1 and toll-like receptor (TLR)2 were decreased in TNF-α-activated HUVEC. CONCLUSION These data show an inhibitory effect of teuclatriol on NF-κB signaling at doses of 312µM and higher, validating the traditional use of Salvia mirzayanii in the treatment of inflammatory diseases. Future work on the mode of action of teuclatriol may provide new lead structures with NF-κB inhibiting properties, lacking possible side effects mediated via alkylating centers of sesquiterpene lactones.
Collapse
Affiliation(s)
- Akram Ziaei
- Department of Immunology, Dr. Sheikh Educational, Research and Treatment Hospital, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jessica Hoppstädter
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| | - Alexandra K Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| | - Mohammad Ramezani
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Amirghofran
- Immunology Department, Autoimmune Disease Research Center, Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Britta Diesel
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| |
Collapse
|
250
|
Pujari R, Kumar N, Ballal S, Eligar SM, Anupama S, Bhat G, Swamy BM, Inamdar SR, Shastry P. Rhizoctonia bataticola lectin (RBL) induces phenotypic and functional characteristics of macrophages in THP-1 cells and human monocytes. Immunol Lett 2015; 163:163-72. [DOI: 10.1016/j.imlet.2014.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 12/06/2014] [Accepted: 12/22/2014] [Indexed: 12/13/2022]
|